SlideShare une entreprise Scribd logo
1  sur  8
Télécharger pour lire hors ligne
Identifying Factors of Microparticles Modified with Arginine
Derivatives That Induce Phenotypic Shifts in Macrophages
Alexander J. Dunn-Sale†
and Kaitlin M. Bratlie*,†,‡,§
†
Department of Chemical & Biological Engineering and ‡
Department of Materials Science & Engineering, Iowa State University,
Ames, Iowa 50011, United States
§
Ames National Laboratory, Ames, Iowa 50011, United States
*S Supporting Information
ABSTRACT: Macrophages are key players in the progression of many diseases,
ranging from rheumatoid arthritis to cancer. Drug delivery systems have the
potential not only to transport payloads to diseased tissue but also to influence
cell behavior. Here, poly(N-isopropylacrylamide-co-acrylic acid) (pNIPAm-co-
AAc) microparticles were modified with 14 different arginine derivatives. These
particles were then incubated with interleukin-4 or lipopolysaccharide-stimulated
macrophages or naïve macrophages (RAW 264.7). The phenotypic state of the
macrophages was assessed by measuring arginase activity, tumor necrosis factor-α
(TNF-α) secretion, and nitrite production. Partial least-squares analysis revealed
material properties and descriptors that shifted the macrophage phenotype for the
three cell conditions in this study. Material descriptors relating to secondary
bonding were suggested to play a role in shifting phenotypes in all three
macrophage culture conditions. These findings suggest that macrophage
responses could be altered through drug delivery vehicles, and this method could be employed to assist in screening potential
candidates.
KEYWORDS: macrophage phenotypes, TNF-α, polymer properties
1. INTRODUCTION
Material properties are known to alter cellular responses.1−8
In
particular, surface modifications can alter cytokine and
molecular secretion profiles.6,8−11
In the case of macrophages,
changes in the amount of secreted molecules can signal a shift
in their phenotype. Macrophages exist on a spectrum, with one
end being pro-inflammatory, denoted M1, and the other being
pro-angiogenic, termed M2. Alternatively, these cells can be
thought of as occupying a color wheel, as proposed by Mosser
and Edwards,12
in which the different activations overlap with
each other, blending into “shades” of macrophage phenotypes.
Newer nomenclature has been proposed by Murray et al.13
to
more accurately characterize the in vitro state of the
macrophage in which the state is characterized by the molecular
activator. The pro-angiogenic macrophages can be activated by
exposure to interleukin-4, M(IL-4), and the pro-inflammatory
cells can be achieved through incubation with lipopolysacchar-
ide M(LPS). The exact material properties that promote
polarization to specific phenotypes remain poorly defined.
Tumor-associated macrophages (TAMs) can exist anywhere
on the spectrum between M1- and M2-like macrophages.
However, they are typically polarized toward an M2-like state.14
The phenotype is currently thought to change from an M1
state during cancer initiation to the predominant M2
phenotype during tumor progression,15,16
creating a permissive
environment for the tumor that encourages angiogenesis,
matrix remodeling, and metastasis. Altering the phenotype of
these cells can be achieved in vitro and in vivo through protein
delivery. Systemic delivery of proteins is generally not feasible
due to short half-lives and a low maximum tolerated dose of
500 ng/kg in the case of IL-12.17
These high doses led to the
development of a local delivery system consisting of the protein
encapsulated in biodegradable polylactic acid to polarize M2
TAMs to an M1 phenotype.18,19
On the other side of this coin,
excessive M1-like macrophage presence is implicated in
inflammatory diseases like arthritis.20,21
Using microspheres
to alter macrophage phenotype in combination with delivering
chemo- or immunochemotherapies may improve outcomes.
Arginine is generally metabolized by two enzymes: nitric
oxide synthase (NOS), which produces citrulline and reactive
nitrogen intermediates, and arginase, which produces ornithine
and urea.22,23
These enzymes are differently up-regulated in
polarized mouse macrophages, with NOS being dominant in
M1 cells and arginase in M2 macrophages.24
Many researchers
used the arginase/NOS ratio to describe the phenotypic
population of macrophages.25−28
Several of the intermediate
products in the conversion of arginine to nitric oxide or urea
have been linked to diseases related to inflammation, which
implicate macrophages in these diseases. Cardiomyopathy29
has
been associated with carnitine deficiency. Supplementation with
Received: January 22, 2016
Accepted: May 13, 2016
Published: May 13, 2016
Article
pubs.acs.org/journal/abseba
© 2016 American Chemical Society 946 DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
creatine has improved outcomes for patients with Huntington’s
disease30
and type 2 diabetes.31
At the other end of the
spectrum, nitroarginine exhibits neuroprotective effects in
Parkinson’s disease.32
These are molecules that are inter-
mediate products in the conversion of arginine to nitric oxide
or urea. Theses diseases are related to inflammation, which
implicates macrophages in these diseases. Using arginine and its
derivatives to alter the phenotypic profile of macrophages has
not been explored, particularly in using these molecules as
surface modifiers.
In this study, we have examined the effect of arginine
derivatives as polymer modifiers in macrophage response.
Arginine is known to have a role in immune responses.22
Furthermore, several of its derivatives are known to impact
NOS production and function. Both 2-amino-3-guanidinopro-
pionic acid and nitroarginine inhibit NOS.33,34
Carnitine
suppresses the production of NO and the protein expression
of inducible nitric oxide synthase (iNOS).35
In addition to
these molecules, several products in arginine metabolism were
studied, including creatine, which is produced through
arginine/glycine amidinotransferase22,23
and citrulline. Other
arginine derivatives are thought to be pro-angiogenic, such as 3-
guanidinopropionic acid, which shifts mitochondrial metabo-
lism from a glycolytic to oxidative mechanism,36
and
acetylcarnitine, which protects against oxidative stress gen-
erated by mitochondrial metabolism.37
Acetylcarnitine has also
been used to decrease iNOS protein expression in multiple
sclerosis patients when delivered orally.38
Additional arginine
derivatives were explored to examine how these modifiers alter
macrophage responses. In addition to M(LPS) and M(IL-4)
cells, naïve macrophages were also tested. Correlations between
the molecules detected and material properties were examined
and used to determine what parameters promote different
macrophage phenotypes under different activation conditions.
2. MATERIALS AND METHODS
Fourteen chemically unique functional groups were coupled to
polymers to assess how they affect macrophage activation. All
experiments had at least four replicates, and error bars indicate the
standard deviation. All materials were purchased from Sigma (St.
Louis, MO) and used as received unless otherwise indicated. Fresh
deionized water (Milli-Q, Barnstead Nanopure, Thermo Scientific,
Waltham, MA) was used throughout this study.
Modifiers that were coupled to p(NIPAm-co-AAc) particles were 2-
amino-3-guanidinopropionic acid, 3-guanidinopropionic acid, nitro-
arginine, creatine (Fisher, Pittsburgh, PA), carnitine, citrulline, 5-
hydroxylysine, acetylglutamine, N-carbamyl-α-aminoisobutyric acid,
acetylcarnitine, 2,4-diaminobutyric acid, acetylornithine, albizziin, and
arginine (Amresco, Solon, OH).
2.1. Polymer Synthesis. 2.1.1. p(NIPAm-co-AAc) Particle Syn-
thesis. Modification of p(NIPAm-co-AAc) particles and their synthesis
have been described previously.5,8
Briefly, NIPAm (2.4 g), N,N-
methylenebis(acrylamide) (0.16 g), and 157 μL of AAc (J.T. Baker,
Center Valley, PA) were dissolved in 100 mL of H2O and stirred
under N2 in a 250 mL round-bottom flask for 30 min at 70 °C. Next,
15 mL of 13.3 mg/mL K2S2O8 was added to the flask. The reaction
was allowed to proceed for 3.5 h, after which the suspension was
slowly cooled to room temperature, filtered with P5 grade filter paper,
and dialyzed for 48 h in Milli-Q water. The particles were freeze-dried
using a lyophilizer (Labconco, Kansas City, MO, 4.5 L).
In a 15 mL tube, 7.5 mL of phosphate buffered saline (PBS, diluted
from a 10× solution, Fisher Scientific, to 0.1 M, pH 7.4), 16.7 μL of
ethylenediamine (Eda), 1.5 mL of 5% w/v p(NIPAm-co-AAc)
particles, and 75 mg of 1-ethyl-3(3-(dimethylamino)propyl)-
carbodiimide hydrochloride (EDC) were vortexed and incubated
overnight at room temperature. The particles were dialyzed against
H2O and lyophilized. The modifiers in Figure 1 were conjugated to the
Eda−p(NIPAm-co-AAc) particles through the carboxylic acid group
on the modifiers in Figure 1 by combining 10 mg of Eda−p(NIPAm-
co-AAc) particles with 2 mg of modifiers and 10 mg of EDC in 2 mL of
PBS at room temperature and stirring overnight. The particles were
dialyzed against H2O and lyophilized.
2.1.2. ζ-Potential. Milli-Q water was neutralized to pH 7 with HCl
or NaOH to ensure that the ions in water would not interfere with the
ζ-potential of particles. A low salt concentration was confirmed by
measuring conductivities at <0.2 mS/cm. A 100 μL aliquot of 1% w/v
of particles was added into 5 mL of water, and ζ-potential was
measured with a Zetasizer Nano Z (Malvern).
2.2. Cell Viability. RAW 264.7 macrophages were cultured at 37
°C with 5% CO2 in 10% fetal bovine serum, 100 U/L penicillin, and
100 μg/L streptomycin in Dulbecco’s modified Eagle’s medium
(DMEM high glucose; Thermo Scientific), to be referred to as
complete medium (CM). RAW 264.7 cells were seeded in a 24-well
plate at 1.25 × 105
cell/cm2
in the presence of 5 μg/mL LPS39,40
or 25
ng/mL IL-441
(eBioscience Inc., San Diego, CA) and incubated 24 h at
37 °C in 5% CO2. A control set of experiments was not activated. After
activation, the medium was replaced with fresh CM, and particles were
added to the wells (100 particles per cell). The cells were incubated for
an additional 24 h. All particles were sterilized by washing three times
in 70% ethanol, followed by three washes in sterile H2O and
centrifugation at 10 000g for 10 min. In a control experiment, cells
were incubated without particles. A control of particle and IL-4 or LPS
in the absence of cells was also conducted. The medium in each well
was collected and stored at −20 °C.
Figure 1. Chemical structures of all molecules used for the modification of p(NIPAm-co-AAc) particles. The letters are used as labels in the following
figures for convenience.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
947
A (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
(MTT) assay was used to determine cell viability in the presence of
the modified particles. A total of 50 μL of MTT (5 mg/mL in DI
H2O) and 500 μL of CM was added to each well, and the plate was
incubated for 2 h at 37 °C in 5% CO2, after which time 425 μL of
media was aspirated. The resulting formazan crystals were dissolved in
500 μL of dimethylsulfoxide (DMSO, Fisher, Pittsburgh, PA). The
absorbance was measured at 540 nm with a reference at 690 nm using
a plate reader (BioTek Synergy HT Multidetection Microplate Reader,
Winooski, VT).
2.3. Measurement of Tumor Necrosis Factor-α Production.
Tumor necrosis factor-α (TNF-α) present in the LPS, IL-4, or naïve
cell supernatant was determined by commercially available immuno-
assay kits (eBioscience Inc., San Diego, CA) and performed as
described by the manufacturer.
2.4. Arginase Activity. After 24 h incubation with particles in
section 2.2, cells were washed with 500 μL of PBS. In each well, 100
μL of cell lysis buffer (150 μL protease inhibitor cocktail (Amresco,
Solon, OH) and 15 μL of Triton X-100 (Acros Organics) filled to 15
mL with DI water) was added, and the plates were incubated on ice for
10 min. Next, 25 μL of the cell lysate was transferred to a 96-well plate,
and 25 μL of 10 mM MnCl2 (Fisher) and 50 mM Tris (Fisher) was
added to each well. Incubation at 55 °C for 10 min activated the
arginase. A solution of 50 μL of 1 M arginine (pH 9.7) was added to
each well, and the plate was incubated at 37 °C for 20 h. The activity
of arginase was determined by measuring the amount of arginine
converted to urea. In 96-well plates, 5 μL of either the urea-containing
samples or standards was added to 200 μL of a 1:2 mixture of solution
1 (1.2 g of o-phthaldialdehyde (Alfa Aesar, Ward Hill, MA), 1 L of
H2O, and 500 μL of HCl (Fisher, Pittsburgh, PA)) and solution 2 (0.6
g of N-(napthyhl)ethylenediamine dihydrochloride (Acros Organics),
5 g of boric acid (Fisher, Pittsburgh, PA), 800 mL of H2O, and 111 mL
of sulfuric acid (Fisher, Pittsburgh, PA), diluted to 1 L with H2O).42
The plates were incubated for 30 min, and the absorbance in each well
was measured at 540 nm with a reference at 630 nm using a plate
reader.
2.5. Griess Reagent Assay. Nitrite concentration in the
supernatant collected in section 2.2 was quantified through a Griess
reagent assay. A standard curve was generated through serial dilutions
of 100 μM NaNO2. To each well were added 150 μL of sample or
standard, 130 μL of DI H2O, and 20 μL of Griess reagent. The plate
was incubated for 20 min, and the absorbance of each well was
measured with a plate reader at 448 nm with a reference of 690 nm.
TNF-α, arginase, and nitrite concentrations were determined at the
same time point of 24 h after introduction of the particles.
2.6. Statistics and Data Analysis. Statistical analysis was
performed using XLSTAT statistical software (New York, NY).
Statistical significance of the mean comparisons was determined by a
two-way ANOVA. Pair-wise comparisons were analyzed with Tukey’s
honest significant difference test. Differences were considered
statistically significant for p < 0.05. The macrophage responses
measured above were analyzed by performing partial least-squares
(PLS) regression on the material descriptors shown in Table 1. These
descriptors were based on descriptors defined by Bicerano.43
3. RESULTS AND DISCUSSION
3.1. Synthesis, ζ-Potential, and Cell Viability. Function-
alized p(NIPAm-co-AAc) particles were synthesized and
modified with the molecules shown in Figure 1 through the
carboxylic acid on the modifier. The ζ-potential of the modified
and unmodified p(NIPAm-co-AAc) is in Figure 2. There was a
range in ζ-potentials for the particles from −4.2 to −20.8 mV.
Cytocompatibility of these materials was tested through an
MTT viability assay. Macrophages were activated with LPS or
IL-4 to simulate M1- and M2-like cells. After being incubated
for 24 h, the medium was replaced and the functionalized
particles were added to each well. Metabolic activity of the cells
was assessed after an additional 24 h incubation period. Naïve
cells, denoted M(0), were also assessed. The results of the
viability assay are shown in Figure 3. All tested conditions were
>70% viable, with most particles and activation conditions
resulting in 90% viability of the macrophages. The largest
decrease in viability came from B, D, G, I, and J for M(LPS), H
for M(IL-4), and L for M(0).
Table 1. Thirty-Eight Molecular Descriptors for the Surface
Modifications Used in This Studya
ID
structure
descriptor
descriptions
1 R freely rotating bonds
2 HD H-bond donors
3 HA H-bond acceptors
4 Nsp
2
number of sp2
carbon atoms
5 N1°C Number of 1° carbon atoms
6 1
X connectivity index 1, calculated based on the molecular graph
of the modifier
7 1
Xv
connectivity index 2, calculated based on the electronic
configuration of the molecular graph of the modifier
8 0
X atomic index 1, calculated based on the number of non-
hydrogen atoms to which a non-hydrogen atom is bound
9 0
Xv
atomic index 2, calculated based on the electronic
configuration of each non-hydrogen atom
10 1
ξ
intensive connectivity index 1, ξ = N
1 X1
11 1
ξv
intensive connectivity index 2, ξ =
ν
ν
N
1 X1
12 0
ξ
intensive atomic index 1, ξ = N
0 X0
13 0
ξv
intensive atomic index 2, ξ =
ν
ν
N
0 X0
14 N number of non-hydrogen atoms
15 NC number of carbon atoms
16 NH number of hydrogen atoms
17 NO number of oxygen atoms
18 NN number of nitrogen atoms
19 NCH2 number of CH2
20 NK NK = 5Namide + 4Nhydroxyl − 3Nether − 5NCC + 3Nsulfone
21 Ndc Ndc = 19NN + 12N(side group O, −S−) + 52Nsulfone − 14Ncyc
22 Ngroup Ngroup = 12Nhydroxyl + 12Namide + 2N(non‑amide −(NH)− unit) −
N(alkyl ether −O−) − NCC + 4N(non‑amide −(CO)− next to a nitrogen)
+ 7N(−(CO)− in carboxylic acid, ketone, or aldehyde) +
2N(other −(CO)−)
23 NMV NMV = −5Nsulfone + 3Nester + 5NCC − 11Ncyc
24 NHη NHη = −7Namide + 6Nether + 5Nsulfone +
3N(six‑membered nonterminal aromatic rings) + 8NCC + 4 N4° carbon atoms
+ 2N3° carbon atoms
25 NUR NUR = −2NOH − 3Ncyc + 4N(six‑membered aromatic rings)
26 Nds Nds = 2NCH2
− 7NCC + 3N(six‑membered aromatic rings) − 12NCO2
27 NPS NPS = −3Nsp
3
carbon atoms + 6Ncarbonyl groups
28 NvdW NvdW = Nmenonar
b
+ Nalamid
c
+ NOH − 4Ncyc + 2NCC
29 Vw Vw = 3.861803°X + 13.7484351
Xv
, van der Waals volume
30 ξNOH ξ =
+ −N N N
NNOH
0.125N O H
31 M molecular weight
32 α polarizability
33 V̅ molar volume
34 log p octanol−water partition ratio
35 log D
(pH 5.5)
distribution coefficient at pH 5.5
36 log D
(pH 7.4)
distribution coefficient at pH 7.4
37 ΔHv enthalpy of vaporization
38 γ surface tension
a
The structure descriptors are defined by Bicerano.43 b
Nmenonar is the
number of methyl groups attached to nonaromatic atoms. c
Nalamid is
the total number of linkages between amide and nonaromatic atoms.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
948
3.2. TNF-α Secretion from Activated and Naïve
Macrophages. Activated and naïve macrophages were
exposed to functionalized p(NIPAm-co-AAc) particles, and
the concentration of secreted TNF-α was measured (Figure
4A). LPS was used to stimulate that M1-like phenotype. The
control in which no particles were added to the cells contained
4.03 ± 0.51 ng/mL. This was statistically higher (p < 0.05) than
the unmodified p(NIPAm-co-AAc) particles and the particles
modified with ethylenediamine of 2.49 ± 0.29 and 2.55 ± 0.25
ng/mL, respectively. Compared to both the unmodified and
Eda-modified particles, modifications D and I−M resulted in
statistically higher TNF-α secretion (p < 0.05). Of these,
albizziin increased TNF-α secretion ∼2-fold. Modifications B,
E, G, and H decreased the TNF-α production (p < 0.05). The
most significant decrease was observed for 3-guanidinopro-
pionic acid of ∼40%. Details of multiple comparisons for these
data are in Table S1.
To explore the M2-like phenotype, macrophages were
stimulated with IL-4. The control without particles resulted
in 2.69 ± 0.08 ng/mL TNF-α measured in the supernatant.
The unmodified p(NIPAm-co-AAc) and Eda-modified particles
resulted in a decrease in TNF-α by 1.78 ± 0.13 and 1.68 ± 0.12
ng/mL, respectively (p < 0.05), similar to the M(LPS) cells.
Modifications A, I−K, and N resulted in increased TNF-α
secretion (p < 0.05), while modifications B, C, E, F, and H
resulted in a decrease (p < 0.05) compared to the unmodified
and Eda particles. Modifications C−G and K resulted in
increases in TNF-α secretion, and particles modified with I, J,
L, and M decreased the TNF-α concentration detected
compared to the unmodified and Eda particles. 2-Amino-3-
guanidinopropionic caused the largest increase in TNF-α
secretion of ∼200%, and 3-guanidinopropionic acid caused the
most significant decrease of ∼40%. Details of multiple
comparisons for these data are in Table S2.
Macrophages without activation were also incubated with the
modified particles, and the secreted TNF-α was assessed. The
control, unmodified p(NIPAm-co-AAc), and Eda particles were
Figure 2. ζ-Potentials for modified p(NIPAm-co-AAc) particles. Data
represent the mean value of three replicates for each sample ±
standard deviation. Eda = ethylenediamine, pN = unmodified
p(NIPAm-co-AAc).
Figure 3. Viability assay showing the cytotoxicity of all particles with
LPS or IL-4 activated or naïve RAW 264.7 cells. The % viability was
normalized to RAW 264.7 cells treated with LPS, IL-4, or naïve cells
without particles. Data represent the mean value of four replicates for
each sample ± standard deviation. Eda = ethylenediamine, pN =
unmodified p(NIPAm-co-AAc).
Figure 4. Molecular expression of (A) TNF-α and (B) urea/nitrite by
macrophages in respose to functionalized p(NIPAm-co-AAc) particles
and stimulation with LPS or IL-4. Naïve cells are also shown. Data
represent the mean value of four replicates for each sample ± standard
deviation. Eda = ethylenediamine, pN = unmodified p(NIPAm-co-
AAc), PC = positive control of cells without materials.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
949
not statistically different with 1.04 ± 0.11, 1.31 ± 0.11, and 1.12
± 0.13 ng/mL TNF-α detected. The largest increase in TNF-α
was found for modifications F and K, with an increase of
∼350%. The largest decrease was observed for modifications I,
J, and L, resulting in an 80% decrease in TNF-α compared with
the unmodified or Eda particles. Details of multiple
comparisons are in Table S3.
Taken together, modifications I, J, and K lead to increases in
TNF-α for both M(LPS) and M(IL-4), while modifications B,
E, and H lead to decreases in TNF-α secretion. The most
dramatic decreases in TNF-α secretion for both M(LPS) and
M(IL-4) were in the presence of the 3-guanidinopropionic acid
modification, which reduced TNF-α secretion by ∼40%
compared to unmodified and Eda-modified particles.
3.3. Urea/Nitrite for Activated and Naïve Macro-
phages. Arginase activity was measured by exposing cell
lysate to arginine and assessing its conversion to urea after 20 h
incubation. Increased arginase activity is suggestive of an M2-
like phenotype. Increased iNOS indicates an M1-like
phenotype. The activity of iNOS was indirectly measured
through a Griess assay in which nitrites, the stable form of NO,
are quantified. NO results from reactive nitrogen intermediates,
which are synthesized by iNOS. Arginine is generally
metabolized through one of these pathways, thus the ratio of
urea/nitrite can assess the extent of polarization toward either
phenotype.25,26
These results are shown in Figure 4B. The
nitrite and urea concentrations are shown as Figure S1A,B.
M(IL-4) cells were found to have 447 ± 68 mg/μmol urea/
nitrite. Cells incubated with A−D and I−L increased the
amount of urea/nitrite measured (p < 0.05), while particles
modified with E−H decreased the levels of urea/nitrite (p <
0.05). Measured levels of urea/nitrite ranged from 252 to 1023
mg/μmol. There was no statistical difference between the
M(IL-4) cells in the absence of particles and M(LPS) cells
incubated with unmodified and Eda-modified p(NIPAm-co-
AAc) particles: 421 ± 100 and 366 ± 70 mg/μmol urea/nitrite,
respectively. Details of multiple comparisons for urea/nitrite for
M(IL-4) are in Table S4.
M1-like cells were also studied using M(LPS)-stimulated
cells. No statistical differences were observed between the
control and cells incubated with unmodified and Eda-modified
particles: 62 ± 5, 69 ± 6, and 70 ± 7 mg/μmol urea/nitrite,
respectively. Particles E−H, K, and M resulted in elevated urea/
nitrite levels. Only particles A and I decreased urea/nitrite (p <
0.05) compared to the positive control, unmodified, and Eda-
modified particles. Details of multiple comparisons for urea/
nitrite for M(LPS) are provided in Table S5. In comparing
trends between M(LPS) and M(IL-4), only modification K
resulted in increasing urea/nitrite levels and no modifications
were able to decrease urea/nitirite in both M(LPS) and M(IL-
4) cells. It is important to note that these results may be
particular to murine macrophage cells. Human macrophages
transport arginine through system y+
L transporters,44
whereas a
cationic amino acid transporter has been suggested as an
arginine transporter in mice.45
Naïve macrophages were also tested for urea/nitrite levels.
The levels of nitrites detected for all M(0) cells were lower than
the standard curve. Statistical analysis on these values is not
reasonable.
3.4. Material Parameters That Influence Macrophage
Phenotypic Shifts. Secretion of TNF-α in the absence of
particles followed the expected trend, in which the highest
levels were observed for M(LPS), followed by M(IL-4) and
M(0).6,46−48
With the exception of modification A, M(LPS)
cells secreted more TNF-α than the M(IL-4) cells when they
were incubated with particles. The M(0) cells exhibited a range
of responses to the particles, secreting less TNF-α in the
presence of modifications A, C, J, Eda, and unmodified
p(NIPAm-co-AAc) than M(LPS) cells. Increased TNF-α was
observed for modification M compared to M(LPS) cells. The
increase in TNF-α secretion may be a result of LPS tolerance
developed in the M(LPS) cells, which leads to a decreased
production of TNF-α.49
Another possibility is that IL-10, a
suppressor of TNF-α, may be secreted. Previous findings have
demonstrated a decrease of TNF-α secretion in M(LPS) cells
by a factor of 21.4 ± 2.5 in the presence of 0.1 ng/mL IL-10.50
Arginase/iNOS is a measure of macrophage polarization,25,26
with high levels denoting M2-like states and low levels
indicating a more M1-like state. Here, we measure this using
urea/nitrite levels. In the case of M(IL-4), both the control and
the cells incubated with particles resulted in higher urea/nitrite
levels, which is expected.
The aim of this work was to identify material properties that
influence M1- and M2-like phenotypes. The trends between
material properties and cell responses were quantitatively
modeled using material descriptors listed in Table 1. These
descriptors were based largely on structural descriptors43
along
with physiochemical properties. PLS was performed on the
measured data using principle component analysis values of
Figure 4A,B as a function of the molecular descriptors in Table
1. The results of the model are presented in Figure 5, where the
horizontal axis represents TNF-α secretion and the vertical axis
is urea/nitrite. Cells that secrete high levels of TNF-α and
exhibit low levels of urea/nitrite are M1-like, while the converse
are M2-like. Both of these regions are outlined in Figure 5. In
addition, the specific descriptors that promote either of these
two extremes are labeled on the plots. For M(IL-4) cells
(Figure 5A), increasing H-bond donors promoted a shift
toward an M1-like phenotype, while increasing H-bond
acceptors, Ndc, log D (5.5), and log D (7.4) retained the M2-
like phenotype. Log D is the octanol−water distribution
coefficient, which can be measured at any pH, and Ndc is a
correction factor for the dielectric constant. It bears mentioning
that the inverse of these material descriptors would result in a
shift toward the opposite phenotype. Based on these
parameters, polarity of the molecule appears to be an important
factor in altering M(IL-4) polarization. It should be noted that
surface tension had a negligible effect on either TNF-α or urea/
nitrite.
A larger number of material properties were identified as
altering M(LPS), shown in Figure 5B. Increasing the number of
freely rotating bonds, aliphatic carbons, van der Waals
interactions (calculated using NνdW), the number of carbon
and hydrogen atoms, and the enthalpy of vaporization
promotes an M2-like phenotype. On the other end of the
spectrum, primary carbon atoms, the number of sp2
carbon
atoms, NMVa correction term for molar volume, NURa
correction term for molar Rao function, ξNOHa correction
term for thermal conductivity, log p, log D (7.4), and log D
(5.5) promoted the M1-like phenotype. All of these parameters
suggest that hydrophobicity plays a dominant role in altering
M(LPS) cells.
Since the levels of nitrites detected for all M(0) cells were
lower than the standard curve, material properties influencing
phenotypic shifts were not analyzed because statistical analysis
on these values is not reasonable.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
950
Interesting similarities exist for the material properties that
promote phenotypic shifts in all the conditions examined here.
Descriptors related to hydrophobicity, van der Waals
interactions, and H-bonding were found to be related to
macrophage polarization for M(LPS) and M(IL-4). The
conventional dogma surrounding ligand−receptor binding
involves all of these interaction forces additively combining to
stabilize the ligand−receptor pair.51
Very likely, serum proteins
present in the culture media adsorb in different configurations
on the particle surface, thus enabling different interactions with
surface receptors. The complexity of macrophage activation and
reprogramming through surface receptors prevents predicting
which receptors these particles are interacting with; however,
examining the impact of material descriptors on altering
macrophage response has the potential to improve material
systems for implanted biomaterials as well as drug delivery
systems.
Based on the observations described above and in Figure 5,
three molecules were selected to alter the macrophage
phenotype: lysine, ornithine, and ureidopropionic acid. These
molecules were conjugated to Eda-modified particles as
described in section 2.1.1. The concentration of secreted
TNF-α and urea/nitrite in response to incubation with the
modified particles is shown in Figure 6.
Ureidopropionic acid was predicted to promote urea/nitrite
and TNF-α secretion in M(IL-4) cells and decrease secretion in
M(LPS). Factors that would increase urea/nitrite and TNF-α
secretion in M(IL-4) cells would be increased distribution
coefficients at pH 5.5 and 7 and a reduction in H-bond
acceptors. Compared to albizziin, ureidopropionic acid has
fewer H-bond acceptors and higher distribution coefficients at
pH 5.5 (−4.0 vs −2.62, respectively). The distribution
coefficients at pH 7 are decreased for ureidopropionic acid
(−4.4 vs −4.01). The distribution coefficients at pH 5.5 and 7
are very similar when compared to those of albizziin to
citrulline, ergo an increase in urea/nitrite and TNF-α secretion
would be expected for ureidopropionic acid compared to
citrulline, as is observed in Figure 5A,B, particularly since
citrulline has an additional H-bond donor compared to
ureidoprionic acid, much like albizziin. In the case of M(LPS)
cells, factors that cause cells to secrete decreased levels of urea/
nitrite are the distribution coefficients at pH 5.5 and 7. In the
Figure 5. Analysis of molecular descriptors to identify polymer
characteristics that promote M1- or M2-like profiles. Pearson’s
correlation between molecular descriptors that promote urea/nitrite
(y-axis) and TNF-α (x-axis) for (A) IL-4 and (B) LPS. Relevant
descriptors that promote M1- or M2-like responses are labeled on the
figure.
Figure 6. Molecular expression of (A) TNF-α and (B) urea/nitrite by
macrophages in respose to functionalized p(NIPAm-co-AAc) particles
and stimulation with LPS or IL-4. Naïve cells are also shown. Data
represent the mean value of four replicates for each sample ± standard
deviation.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
951
case of M(LPS) cells, the higher distribution coefficient for
ureidopropionic acid compared to that of either albizziin or
citrulline would cause a decrease in the urea/nitrite and TNF-α
levels, as was observed. The number of freely rotating bonds is
also lower for ureidopropionic acid compared to albizziin and
citrulline.
The difference between lysine and ornithine is an additional
aliphatic CH2 group in lysine, which also results in an additional
freely rotating bond for lysine. The other parameters
influencing urea/nitrite production are similar for the two
modifiers. This resulted in predictions that lysine would result
in increased nitrite/urea secretion compared to ornithine. The
additional CH2 group results in a 2-fold increase in urea/nitrite
for the two modifiers in the case of M(LPS) cells. In examining
the response of M(IL-4) cells, no difference between lysine-
and ornithine-modified particles was expected for urea/nitrite
and TNF-α levels since the differences in their structures did
not result in differences in the material descriptors identified in
Figure 5A,B.
4. CONCLUSIONS
In summary, we have examined how a library of 14 surface
modifiers alters macrophage response and have correlated those
responses to material descriptors that promote shifts in
phenotypes. Similarities between all three macrophage treat-
ment conditions exist in that intermolecular nonbonding
interactions play an important role in altering macrophage
responses. The differences between these three conditions
suggest the possibility that material properties can be exploited
to manipulate macrophage responses, possibly reprogramming
these cells without exogenous protein delivery. Semiquantita-
tive methods such as those employed in this study have the
potential to both improve screening for materials that promote
M1- or M2-like phenotypes in macrophages and add to the
current library of knowledge pertaining to how materials
influence cellular responses.
■ ASSOCIATED CONTENT
*S Supporting Information
The Supporting Information is available free of charge on the
ACS Publications website at DOI: 10.1021/acsbiomater-
ials.6b00041.
Details of multiple comparisons for TNF-α for M(LPS),
M(IL-4), and M(0) cells and for urea/nitrite for M(LPS)
and M(IL-4) cells; nitrite and urea measured for
M(LPS), M(IL-4), and M(0) cells (PDF)
■ AUTHOR INFORMATION
Corresponding Author
*Tel: 515-294-7304. Fax: 515-294-5444. E-mail: kbratlie@
iastate.edu.
Notes
The authors declare no competing financial interest.
■ ACKNOWLEDGMENTS
This work was supported by the National Science Foundation
under Grant No. CBET 1227867 and the Roy J. Carver
Charitable Trust Grant No. 13-4265. The authors also
acknowledge support from NSF ARI-R2 (CMMI-0963224)
for funding the renovation of the research laboratories used for
these studies.
■ REFERENCES
(1) Acharya, A. P.; Dolgova, N. V.; Clare-Salzler, M. J.; Keselowsky,
B. G. Adhesive substrate-modulation of adaptive immune responses.
Biomaterials 2008, 29, 4736−50.
(2) Schutte, R. J.; Xie, L.; Klitzman, B.; Reichert, W. M. In vivo
cytokine-associated responses to biomaterials. Biomaterials 2009, 30,
160−8.
(3) Anderson, J. M.; Rodriguez, A.; Chang, D. T. Foreign body
reaction to biomaterials. Semin. Immunol. 2008, 20, 86−100.
(4) Nilsson, B.; Ekdahl, K. N.; Mollnes, T. E.; Lambris, J. D. The role
of complement in biomaterial-induced inflammation. Mol. Immunol.
2007, 44, 82−94.
(5) Wang, D.; Phan, N.; Isely, C.; Bruene, L.; Bratlie, K. M. The
effect of polymer chemistry and macrophage phenotype on particle
internalization. Biomacromolecules 2014, 15, 4102−10.
(6) Wang, D.; Bratlie, K. M. Influence of Polymer Chemistry on
Cytokine Secretion from Polarized Macrophages. ACS Biomater. Sci.
Eng. 2015, 1, 166−74.
(7) Bygd, H. C.; Akilbekova, D.; Muñoz, A.; Forsmark, K. D.; Bratlie,
K. M. Poly-l-arginine based materials as instructive substrates for
fibroblast synthesis of collagen. Biomaterials 2015, 63, 47−57.
(8) Bygd, H. C.; Forsmark, K. D.; Bratlie, K. M. Altering in vivo
macrophage responses with modified polymer properties. Biomaterials
2015, 56, 187−97.
(9) Petersen, L. K.; Xue, L.; Wannemuehler, M. J.; Rajan, K.;
Narasimhan, B. The simultaneous effect of polymer chemistry and
device geometry on the in vitro activation of murine dendritic cells.
Biomaterials 2009, 30, 5131−42.
(10) Ariganello, M. B.; Simionescu, D. T.; Labow, R. S.; Lee, J. M.
Macrophage differentiation and polarization on a decellularized
pericardial biomaterial. Biomaterials 2011, 32, 439−49.
(11) Rodriguez, A.; Meyerson, H.; Anderson, J. M. Quantitative in
vivo cytokine analysis at synthetic biomaterial implant sites. J. Biomed.
Mater. Res., Part A 2008, 89, 152−9.
(12) Mosser, D. M. The many faces of macrophage activation. J.
Leukocyte Biol. 2003, 73, 209−12.
(13) Murray, P. J.; Allen, J. E.; Biswas, S. K.; Fisher, E. A.; Gilroy, D.
W.; Goerdt, S.; et al. Macrophage Activation and Polarization:
Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14−
20.
(14) Mantovani, A.; Locati, M. Tumor-associated macrophages as a
paradigm of macrophage plasticity, diversity, and polarization: lessons
and open questions. Arterioscler., Thromb., Vasc. Biol. 2013, 33, 1478−
83.
(15) Biswas, S. K.; Sica, A.; Lewis, C. E. Plasticity of Macrophage
Function during Tumor Progression: Regulation by Distinct Molecular
Mechanisms. J. Immunol. 2008, 180, 2011−7.
(16) Qian, B.-Z.; Pollard, J. W. Macrophage diversity enhances tumor
progression and metastasis. Cell 2010, 141, 39−51.
(17) Gollob, J A.; Mier, J. W.; Veenstra, K.; McDermott, D. F.;
Clancy, D.; Clancy, M.; et al. Phase I trial of twice-weekly intravenous
interleukin 12 in patients with metastatic renal cell cancer or malignant
melanoma: ability to maintain IFN-gamma induction is associated with
clinical response. Clin. Cancer Res. 2000, 6, 1678−92.
(18) Hill, H.; Conway, T.; Sabel, M.; Jong, Y.; Mathiowitz, E.;
Bankert, R.; et al. Cancer immunotherapy with interleukin 12 and
granulocyte-macrophage colony-stimulating factor-encapsulated mi-
crospheres: coinduction of innate and adaptive antitumor immunity
and cure of disseminated disease. Cancer Res. 2002, 62, 7254−63.
(19) Watkins, S. K.; Egilmez, N. K.; Suttles, J.; Stout, R. D. IL-12
rapidly alters the functional profile of tumor-associated and tumor-
inflitrating macrophages in vitro and in vivo. J. Immunol. 2007, 178,
1357−62.
(20) Hofkens, W.; Schelbergen, R.; Storm, G.; van den Berg, W. B.;
van Lent, P. L. Liposomal Targeting of Prednisolone Phosphate to
Synovial Lining Macrophages during Experimental Arthritis Inhibits
M1 Activation but Does Not Favor M2 Differentiation. PLoS One
2013, 8, e54016.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
952
(21) Jain, S.; Tran, T.-H.; Amiji, M. Macrophage repolarization with
targeted alginate nanoparticles containing IL-10 plasmid DNA for the
treatment of experimental arthritis. Biomaterials 2015, 61, 162−77.
(22) Bronte, V.; Zanovello, P. Regulation of immune responses by L-
arginine metabolism. Nat. Rev. Immunol. 2005, 5, 641−54.
(23) Morris, S. M. Arginine metabolism: boundaries of our
knowledge. J. Nutr. 2007, 137, 1602S−1609S.
(24) Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.;
Locati, M. The chemokine system in diverse forms of macrophage
activation and polarization. Trends Immunol. 2004, 25, 677−86.
(25) Lumeng, C. N.; Bodzin, J. L.; Saltiel, A. R. Obesity induces a
phenotypic switch in adipose tissue macrophage polarization. J. Clin.
Invest. 2007, 117, 175−84.
(26) Allen, J. E.; Loke, P. Divergent roles for macrophages in
lymphatic filariasis. Parasite Immunol. 2001, 23, 345−52.
(27) Murphy, B. S.; Sundareshan, V.; Cory, T. J.; Hayes, D.; Anstead,
M. I.; Feola, D. J. Azithromycin alters macrophage phenotype. J.
Antimicrob. Chemother. 2008, 61, 554−60.
(28) Rath, M.; Muller, I.; Kropf, P.; Closs, E. I.; Munder, M.
Metabolism via Arginase or Nitric Oxide Synthase: Two Competing
Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 1−10.
(29) Amat di San Filippo, C.; Taylor, M. R. G.; Mestroni, L.; Botto,
L. D.; Longo, N. Cardiomyopathy and carnitine deficiency. Mol. Genet.
Metab. 2008, 94, 162−6.
(30) Ferrante, R.; Andreassen, O.; Jenkins, B.; Dedeoglu, a;
Kuemmerle, S.; Kubilus, J.; et al. Neuroprotective effects of creatine
in a transgenic mouse model of Huntington’s disease. J. Neurosci. 2000,
20, 4389−97.
(31) GUALANO, B.; DE SALLES PAINNELI, V.; ROSCHEL, H.;
ARTIOLI, G. G.; NEVES, M.; DE SÁ PINTO, A. L.; et al. Creatine in
Type 2 Diabetes. Med. Sci. Sports Exercise 2011, 43, 770−8.
(32) Arimoto, T.; Bing, G. Up-regulation of inducible nitric oxide
synthase in the substantia nigra by lipopolysaccharide causes microglial
activation and neurodegeneration. Neurobiol. Dis. 2003, 12, 35−45.
(33) Grant, S. K.; Green, B. G.; Stiffey-Wilusz, J.; Durette, P. L.;
Shah, S. K.; Kozarich, J. W. Structural requirements for human
inducible nitric oxide synthase substrates and substrate analogue
inhibitors. Biochemistry 1998, 37, 4174−80.
(34) Jang, J. J.; Ho, H. K.; Kwan, H. H.; Fajardo, L. F.; Cooke, J. P.
Angiogenesis is impaired by hypercholesterolemia: role of asymmetric
dimethylarginine. Circulation 2000, 102, 1414−9.
(35) Koc, a; Ozkan, T.; Karabay, a Z; Sunguroglu, a; Aktan, F. Effect
of L-carnitine on the synthesis of nitric oxide in RAW 264·7 murine
macrophage cell line. Cell Biochem Funct 2011, 29, 679−85.
(36) Oudman, I.; Clark, J. F.; Brewster, L. M. The Effect of the
Creatine Analogue Beta-guanidinopropionic Acid on Energy Metab-
olism: A Systematic Review. PLoS One 2013, 8, e52879.
(37) Hagen, T. M.; Ingersoll, R. T.; Wehr, C. M.; Lykkesfeldt, J.;
Vinarsky, V.; Bartholomew, J. C.; et al. Acetyl-L-carnitine fed to old
rats partially restores mitochondrial function and ambulatory activity.
Proc. Natl. Acad. Sci. U. S. A. 1998, 95, 9562−6.
(38) Calabrese, V.; Scapagnini, G.; Ravagna, a.; Bella, R.; Butterfield,
D a.; Calvani, M.; et al. Disruption of Thiol Homeostasis and
Nitrosative Stress in the Cerebrospinal Fluid of Patients with Active
Multiple Sclerosis: Evidence for a Protective Role of Acetylcarnitine.
Neurochem. Res. 2003, 28, 1321−8.
(39) Ricote, M.; Li, A. C.; Willson, T. M.; Kelly, C. J.; Glass, C. K.
The peroxisome proliferator-activated receptor-gamma is a negative
regulator of macrophage activation. Nature 1998, 391, 79−82.
(40) Guastadisegni, C.; Minghetti, L.; Nicolini, A.; Polazzi, E.; Ade,
P.; Balduzzi, M.; et al. Prostaglandin E2 synthesis is differentially
affected by reactive nitrogen intermediates in cultured rat microglia
and RAW 264.7 cells. FEBS Lett. 1997, 413, 314−8.
(41) Motrán, C. C.; Díaz, F. L.; Gruppi, A.; Slavin, D.; Chatton, B.;
Bocco, J. L. Human pregnancy-specific glycoprotein 1a (PSG1a)
induces alternative activation in human and mouse monocytes and
suppresses the accessory cell-dependent T cell proliferation. J.
Leukocyte Biol. 2002, 72, 512−21.
(42) Levinson, S. S. Kinetic centrifugal analyzer and manual
determination of serum urea nitrogen, with use of o-phthaldialdehyde
reagent. Clin. Chem. 1978, 24, 2199−202.
(43) Bicerano, J. Prediction of Polymer Properties; Marcel Dekker Inc.:
New York, 2002.
(44) Rath, M.; Muller, I.; Kropf, P.; Closs, E. I.; Munder, M.
Metabolism via Arginase or Nitric Oxide Synthase: Two Competing
Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 1−10.
(45) Manner, C. K.; Nicholson, B.; MacLeod, C. L. CAT2 arginine
transporter deficiency significantly reduces iNOS-mediated NO
production in astrocytes. J. Neurochem. 2003, 85, 476−82.
(46) Akilbekova, D.; Philiph, R.; Graham, A.; Bratlie, K. M.
Macrophage reprogramming: influence of latex beads with various
functional groups on macrophage phenotype and phagocytic uptake in
vitro. J. Biomed. Mater. Res., Part A 2015, 103, 262.
(47) Barth, K a.; Waterfield, J. D.; Brunette, D. M. The effect of
surface roughness on RAW 264.7 macrophage phenotype. J. Biomed.
Mater. Res., Part A 2013, 101, 2679−88.
(48) Varin, A.; Mukhopadhyay, S.; Herbein, G.; Gordon, S.
Alternative activation of macrophages by IL-4 impairs phagocytosis
of pathogens but potentiates microbial-induced signalling and cytokine
secretion. Blood 2010, 115, 353−62.
(49) Docke, F. R. W.; Bundschuhrt, D. S.; Hartung, T.; Wendel, A.;
Volk, H. In vitro prevention and reversal of lipopolysaccharide
desensitization by IFN-gamma, IL-12, and granulocyte-macrophage
colony-stimulating factor. J. Immunol. 1997, 158, 2911−8.
(50) Bogdan, C.; Vodovotz, Y.; Nathan, C. Macrophage deactivation
by interleukin 10. J. Exp. Med. 1991, 174, 1549−55.
(51) Seong, S.-Y.; Matzinger, P. Hydrophobicity: an ancient damage-
associated molecular pattern that initiates innate immune responses.
Nat. Rev. Immunol. 2004, 4, 469−78.
ACS Biomaterials Science & Engineering Article
DOI: 10.1021/acsbiomaterials.6b00041
ACS Biomater. Sci. Eng. 2016, 2, 946−953
953

Contenu connexe

Tendances

The complement system
The complement systemThe complement system
The complement systemkamilKhan63
 
biomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdfbiomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdfMarcoAntonioRamosIba1
 
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...Farya Mubarik
 
The Complement System
The Complement SystemThe Complement System
The Complement SystemYbeb18
 
Proteome-wide covalent ligand discovery in native biological systems
Proteome-wide covalent ligand discovery in native biological systemsProteome-wide covalent ligand discovery in native biological systems
Proteome-wide covalent ligand discovery in native biological systemsMegha Majumder
 
In silico discovery of dna methyltransferase inhibitors (1)
In silico discovery of dna methyltransferase inhibitors (1)In silico discovery of dna methyltransferase inhibitors (1)
In silico discovery of dna methyltransferase inhibitors (1)angelicagonzalez10
 
Biochemistry tom murray (poster)
Biochemistry tom murray (poster)Biochemistry tom murray (poster)
Biochemistry tom murray (poster)Tommurray111
 
Hayes and Clarkson Biochem J 1982
Hayes and Clarkson Biochem J 1982Hayes and Clarkson Biochem J 1982
Hayes and Clarkson Biochem J 1982George Clarkson
 
The complement system
The complement systemThe complement system
The complement systemAnushi Jain
 
2 complemento receptores - segunda apresentação
2  complemento receptores - segunda apresentação2  complemento receptores - segunda apresentação
2 complemento receptores - segunda apresentaçãoufamimunologia
 
Complement system
Complement systemComplement system
Complement systemPawan Kumar
 
Metabolomics: The Next Generation of Biochemistry
Metabolomics: The Next Generation of BiochemistryMetabolomics: The Next Generation of Biochemistry
Metabolomics: The Next Generation of BiochemistryMargaret Eason
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methileneJorge Parodi
 

Tendances (20)

The complement system
The complement systemThe complement system
The complement system
 
biomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdfbiomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdf
 
Complement final
Complement finalComplement final
Complement final
 
Biotech lett
Biotech lettBiotech lett
Biotech lett
 
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...
The interaction of Nrf2 and Glyoxalase I in response to lipid loading in Hepa...
 
The Complement System
The Complement SystemThe Complement System
The Complement System
 
Lectins
LectinsLectins
Lectins
 
Proteome-wide covalent ligand discovery in native biological systems
Proteome-wide covalent ligand discovery in native biological systemsProteome-wide covalent ligand discovery in native biological systems
Proteome-wide covalent ligand discovery in native biological systems
 
In silico discovery of dna methyltransferase inhibitors (1)
In silico discovery of dna methyltransferase inhibitors (1)In silico discovery of dna methyltransferase inhibitors (1)
In silico discovery of dna methyltransferase inhibitors (1)
 
LSD1 - bmc-paper
LSD1 - bmc-paperLSD1 - bmc-paper
LSD1 - bmc-paper
 
( Isi, 2016) international journal of biological macromolecules gst kappa 201...
( Isi, 2016) international journal of biological macromolecules gst kappa 201...( Isi, 2016) international journal of biological macromolecules gst kappa 201...
( Isi, 2016) international journal of biological macromolecules gst kappa 201...
 
Biochemistry tom murray (poster)
Biochemistry tom murray (poster)Biochemistry tom murray (poster)
Biochemistry tom murray (poster)
 
Hayes and Clarkson Biochem J 1982
Hayes and Clarkson Biochem J 1982Hayes and Clarkson Biochem J 1982
Hayes and Clarkson Biochem J 1982
 
The complement system
The complement systemThe complement system
The complement system
 
2 complemento receptores - segunda apresentação
2  complemento receptores - segunda apresentação2  complemento receptores - segunda apresentação
2 complemento receptores - segunda apresentação
 
Complement system
Complement systemComplement system
Complement system
 
BBL2
BBL2BBL2
BBL2
 
Metabolomics: The Next Generation of Biochemistry
Metabolomics: The Next Generation of BiochemistryMetabolomics: The Next Generation of Biochemistry
Metabolomics: The Next Generation of Biochemistry
 
Complements and complement deficiency
Complements and complement deficiencyComplements and complement deficiency
Complements and complement deficiency
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methilene
 

Similaire à Masters Research (Published)

Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...
Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...
Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...CrimsonpublishersMedical
 
Environmental Factor - July 2014_ Intramural papers of the month
Environmental Factor - July 2014_ Intramural papers of the monthEnvironmental Factor - July 2014_ Intramural papers of the month
Environmental Factor - July 2014_ Intramural papers of the monthXunhai 郑训海
 
Screening and isolation of medicinally important L-asparaginase enzyme from a...
Screening and isolation of medicinally important L-asparaginase enzyme from a...Screening and isolation of medicinally important L-asparaginase enzyme from a...
Screening and isolation of medicinally important L-asparaginase enzyme from a...pharmaindexing
 
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...Ashujit
 
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...Angela Farngren
 
OMICS in Crop Improvement.pptx
OMICS in Crop Improvement.pptxOMICS in Crop Improvement.pptx
OMICS in Crop Improvement.pptxGautham S
 
Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011nemata
 
00046 Ma Hughes 2006 Jpr V5p54
00046 Ma Hughes 2006 Jpr V5p5400046 Ma Hughes 2006 Jpr V5p54
00046 Ma Hughes 2006 Jpr V5p54jcruzsilva
 
Nucl_Acids_Res-2015-Yang-NAR_gkv060
Nucl_Acids_Res-2015-Yang-NAR_gkv060Nucl_Acids_Res-2015-Yang-NAR_gkv060
Nucl_Acids_Res-2015-Yang-NAR_gkv060Bing Yang
 
Seminario biología molecular
Seminario biología molecular Seminario biología molecular
Seminario biología molecular MarianaRestrepo13
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdfHadgi1
 
Central dogma of genetic information
Central dogma of genetic informationCentral dogma of genetic information
Central dogma of genetic informationDaniel Vanegas
 
acs.jmedchem.5b00495
acs.jmedchem.5b00495acs.jmedchem.5b00495
acs.jmedchem.5b00495Justin Murray
 
26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne
 26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne 26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne
26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ neMargaritoWhitt221
 
A mild reduction of food intake slows disease progression in an orthologous m...
A mild reduction of food intake slows disease progression in an orthologous m...A mild reduction of food intake slows disease progression in an orthologous m...
A mild reduction of food intake slows disease progression in an orthologous m...Mina Rezaei
 

Similaire à Masters Research (Published) (20)

Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...
Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...
Crimson publishers-5-MethylcytosineDNA Methylation Patterns among Gut Predomi...
 
Environmental Factor - July 2014_ Intramural papers of the month
Environmental Factor - July 2014_ Intramural papers of the monthEnvironmental Factor - July 2014_ Intramural papers of the month
Environmental Factor - July 2014_ Intramural papers of the month
 
Screening and isolation of medicinally important L-asparaginase enzyme from a...
Screening and isolation of medicinally important L-asparaginase enzyme from a...Screening and isolation of medicinally important L-asparaginase enzyme from a...
Screening and isolation of medicinally important L-asparaginase enzyme from a...
 
824746
824746824746
824746
 
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
 
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...
Inhibition of glutathione by buthionine sulfoximine enhanced the anti-cancer ...
 
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...
Evaluation of the changes in the gene CYP3A4 expression in HepG2 cells under ...
 
pone.0143384
pone.0143384pone.0143384
pone.0143384
 
PROTEOMICS.pptx
PROTEOMICS.pptxPROTEOMICS.pptx
PROTEOMICS.pptx
 
cgt201624a
cgt201624acgt201624a
cgt201624a
 
OMICS in Crop Improvement.pptx
OMICS in Crop Improvement.pptxOMICS in Crop Improvement.pptx
OMICS in Crop Improvement.pptx
 
Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011
 
00046 Ma Hughes 2006 Jpr V5p54
00046 Ma Hughes 2006 Jpr V5p5400046 Ma Hughes 2006 Jpr V5p54
00046 Ma Hughes 2006 Jpr V5p54
 
Nucl_Acids_Res-2015-Yang-NAR_gkv060
Nucl_Acids_Res-2015-Yang-NAR_gkv060Nucl_Acids_Res-2015-Yang-NAR_gkv060
Nucl_Acids_Res-2015-Yang-NAR_gkv060
 
Seminario biología molecular
Seminario biología molecular Seminario biología molecular
Seminario biología molecular
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdf
 
Central dogma of genetic information
Central dogma of genetic informationCentral dogma of genetic information
Central dogma of genetic information
 
acs.jmedchem.5b00495
acs.jmedchem.5b00495acs.jmedchem.5b00495
acs.jmedchem.5b00495
 
26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne
 26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne 26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne
26 ASBMB TODAY FEBRUARY 2021Discovering an old DoGs’ ne
 
A mild reduction of food intake slows disease progression in an orthologous m...
A mild reduction of food intake slows disease progression in an orthologous m...A mild reduction of food intake slows disease progression in an orthologous m...
A mild reduction of food intake slows disease progression in an orthologous m...
 

Masters Research (Published)

  • 1. Identifying Factors of Microparticles Modified with Arginine Derivatives That Induce Phenotypic Shifts in Macrophages Alexander J. Dunn-Sale† and Kaitlin M. Bratlie*,†,‡,§ † Department of Chemical & Biological Engineering and ‡ Department of Materials Science & Engineering, Iowa State University, Ames, Iowa 50011, United States § Ames National Laboratory, Ames, Iowa 50011, United States *S Supporting Information ABSTRACT: Macrophages are key players in the progression of many diseases, ranging from rheumatoid arthritis to cancer. Drug delivery systems have the potential not only to transport payloads to diseased tissue but also to influence cell behavior. Here, poly(N-isopropylacrylamide-co-acrylic acid) (pNIPAm-co- AAc) microparticles were modified with 14 different arginine derivatives. These particles were then incubated with interleukin-4 or lipopolysaccharide-stimulated macrophages or naïve macrophages (RAW 264.7). The phenotypic state of the macrophages was assessed by measuring arginase activity, tumor necrosis factor-α (TNF-α) secretion, and nitrite production. Partial least-squares analysis revealed material properties and descriptors that shifted the macrophage phenotype for the three cell conditions in this study. Material descriptors relating to secondary bonding were suggested to play a role in shifting phenotypes in all three macrophage culture conditions. These findings suggest that macrophage responses could be altered through drug delivery vehicles, and this method could be employed to assist in screening potential candidates. KEYWORDS: macrophage phenotypes, TNF-α, polymer properties 1. INTRODUCTION Material properties are known to alter cellular responses.1−8 In particular, surface modifications can alter cytokine and molecular secretion profiles.6,8−11 In the case of macrophages, changes in the amount of secreted molecules can signal a shift in their phenotype. Macrophages exist on a spectrum, with one end being pro-inflammatory, denoted M1, and the other being pro-angiogenic, termed M2. Alternatively, these cells can be thought of as occupying a color wheel, as proposed by Mosser and Edwards,12 in which the different activations overlap with each other, blending into “shades” of macrophage phenotypes. Newer nomenclature has been proposed by Murray et al.13 to more accurately characterize the in vitro state of the macrophage in which the state is characterized by the molecular activator. The pro-angiogenic macrophages can be activated by exposure to interleukin-4, M(IL-4), and the pro-inflammatory cells can be achieved through incubation with lipopolysacchar- ide M(LPS). The exact material properties that promote polarization to specific phenotypes remain poorly defined. Tumor-associated macrophages (TAMs) can exist anywhere on the spectrum between M1- and M2-like macrophages. However, they are typically polarized toward an M2-like state.14 The phenotype is currently thought to change from an M1 state during cancer initiation to the predominant M2 phenotype during tumor progression,15,16 creating a permissive environment for the tumor that encourages angiogenesis, matrix remodeling, and metastasis. Altering the phenotype of these cells can be achieved in vitro and in vivo through protein delivery. Systemic delivery of proteins is generally not feasible due to short half-lives and a low maximum tolerated dose of 500 ng/kg in the case of IL-12.17 These high doses led to the development of a local delivery system consisting of the protein encapsulated in biodegradable polylactic acid to polarize M2 TAMs to an M1 phenotype.18,19 On the other side of this coin, excessive M1-like macrophage presence is implicated in inflammatory diseases like arthritis.20,21 Using microspheres to alter macrophage phenotype in combination with delivering chemo- or immunochemotherapies may improve outcomes. Arginine is generally metabolized by two enzymes: nitric oxide synthase (NOS), which produces citrulline and reactive nitrogen intermediates, and arginase, which produces ornithine and urea.22,23 These enzymes are differently up-regulated in polarized mouse macrophages, with NOS being dominant in M1 cells and arginase in M2 macrophages.24 Many researchers used the arginase/NOS ratio to describe the phenotypic population of macrophages.25−28 Several of the intermediate products in the conversion of arginine to nitric oxide or urea have been linked to diseases related to inflammation, which implicate macrophages in these diseases. Cardiomyopathy29 has been associated with carnitine deficiency. Supplementation with Received: January 22, 2016 Accepted: May 13, 2016 Published: May 13, 2016 Article pubs.acs.org/journal/abseba © 2016 American Chemical Society 946 DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953
  • 2. creatine has improved outcomes for patients with Huntington’s disease30 and type 2 diabetes.31 At the other end of the spectrum, nitroarginine exhibits neuroprotective effects in Parkinson’s disease.32 These are molecules that are inter- mediate products in the conversion of arginine to nitric oxide or urea. Theses diseases are related to inflammation, which implicates macrophages in these diseases. Using arginine and its derivatives to alter the phenotypic profile of macrophages has not been explored, particularly in using these molecules as surface modifiers. In this study, we have examined the effect of arginine derivatives as polymer modifiers in macrophage response. Arginine is known to have a role in immune responses.22 Furthermore, several of its derivatives are known to impact NOS production and function. Both 2-amino-3-guanidinopro- pionic acid and nitroarginine inhibit NOS.33,34 Carnitine suppresses the production of NO and the protein expression of inducible nitric oxide synthase (iNOS).35 In addition to these molecules, several products in arginine metabolism were studied, including creatine, which is produced through arginine/glycine amidinotransferase22,23 and citrulline. Other arginine derivatives are thought to be pro-angiogenic, such as 3- guanidinopropionic acid, which shifts mitochondrial metabo- lism from a glycolytic to oxidative mechanism,36 and acetylcarnitine, which protects against oxidative stress gen- erated by mitochondrial metabolism.37 Acetylcarnitine has also been used to decrease iNOS protein expression in multiple sclerosis patients when delivered orally.38 Additional arginine derivatives were explored to examine how these modifiers alter macrophage responses. In addition to M(LPS) and M(IL-4) cells, naïve macrophages were also tested. Correlations between the molecules detected and material properties were examined and used to determine what parameters promote different macrophage phenotypes under different activation conditions. 2. MATERIALS AND METHODS Fourteen chemically unique functional groups were coupled to polymers to assess how they affect macrophage activation. All experiments had at least four replicates, and error bars indicate the standard deviation. All materials were purchased from Sigma (St. Louis, MO) and used as received unless otherwise indicated. Fresh deionized water (Milli-Q, Barnstead Nanopure, Thermo Scientific, Waltham, MA) was used throughout this study. Modifiers that were coupled to p(NIPAm-co-AAc) particles were 2- amino-3-guanidinopropionic acid, 3-guanidinopropionic acid, nitro- arginine, creatine (Fisher, Pittsburgh, PA), carnitine, citrulline, 5- hydroxylysine, acetylglutamine, N-carbamyl-α-aminoisobutyric acid, acetylcarnitine, 2,4-diaminobutyric acid, acetylornithine, albizziin, and arginine (Amresco, Solon, OH). 2.1. Polymer Synthesis. 2.1.1. p(NIPAm-co-AAc) Particle Syn- thesis. Modification of p(NIPAm-co-AAc) particles and their synthesis have been described previously.5,8 Briefly, NIPAm (2.4 g), N,N- methylenebis(acrylamide) (0.16 g), and 157 μL of AAc (J.T. Baker, Center Valley, PA) were dissolved in 100 mL of H2O and stirred under N2 in a 250 mL round-bottom flask for 30 min at 70 °C. Next, 15 mL of 13.3 mg/mL K2S2O8 was added to the flask. The reaction was allowed to proceed for 3.5 h, after which the suspension was slowly cooled to room temperature, filtered with P5 grade filter paper, and dialyzed for 48 h in Milli-Q water. The particles were freeze-dried using a lyophilizer (Labconco, Kansas City, MO, 4.5 L). In a 15 mL tube, 7.5 mL of phosphate buffered saline (PBS, diluted from a 10× solution, Fisher Scientific, to 0.1 M, pH 7.4), 16.7 μL of ethylenediamine (Eda), 1.5 mL of 5% w/v p(NIPAm-co-AAc) particles, and 75 mg of 1-ethyl-3(3-(dimethylamino)propyl)- carbodiimide hydrochloride (EDC) were vortexed and incubated overnight at room temperature. The particles were dialyzed against H2O and lyophilized. The modifiers in Figure 1 were conjugated to the Eda−p(NIPAm-co-AAc) particles through the carboxylic acid group on the modifiers in Figure 1 by combining 10 mg of Eda−p(NIPAm- co-AAc) particles with 2 mg of modifiers and 10 mg of EDC in 2 mL of PBS at room temperature and stirring overnight. The particles were dialyzed against H2O and lyophilized. 2.1.2. ζ-Potential. Milli-Q water was neutralized to pH 7 with HCl or NaOH to ensure that the ions in water would not interfere with the ζ-potential of particles. A low salt concentration was confirmed by measuring conductivities at <0.2 mS/cm. A 100 μL aliquot of 1% w/v of particles was added into 5 mL of water, and ζ-potential was measured with a Zetasizer Nano Z (Malvern). 2.2. Cell Viability. RAW 264.7 macrophages were cultured at 37 °C with 5% CO2 in 10% fetal bovine serum, 100 U/L penicillin, and 100 μg/L streptomycin in Dulbecco’s modified Eagle’s medium (DMEM high glucose; Thermo Scientific), to be referred to as complete medium (CM). RAW 264.7 cells were seeded in a 24-well plate at 1.25 × 105 cell/cm2 in the presence of 5 μg/mL LPS39,40 or 25 ng/mL IL-441 (eBioscience Inc., San Diego, CA) and incubated 24 h at 37 °C in 5% CO2. A control set of experiments was not activated. After activation, the medium was replaced with fresh CM, and particles were added to the wells (100 particles per cell). The cells were incubated for an additional 24 h. All particles were sterilized by washing three times in 70% ethanol, followed by three washes in sterile H2O and centrifugation at 10 000g for 10 min. In a control experiment, cells were incubated without particles. A control of particle and IL-4 or LPS in the absence of cells was also conducted. The medium in each well was collected and stored at −20 °C. Figure 1. Chemical structures of all molecules used for the modification of p(NIPAm-co-AAc) particles. The letters are used as labels in the following figures for convenience. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 947
  • 3. A (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to determine cell viability in the presence of the modified particles. A total of 50 μL of MTT (5 mg/mL in DI H2O) and 500 μL of CM was added to each well, and the plate was incubated for 2 h at 37 °C in 5% CO2, after which time 425 μL of media was aspirated. The resulting formazan crystals were dissolved in 500 μL of dimethylsulfoxide (DMSO, Fisher, Pittsburgh, PA). The absorbance was measured at 540 nm with a reference at 690 nm using a plate reader (BioTek Synergy HT Multidetection Microplate Reader, Winooski, VT). 2.3. Measurement of Tumor Necrosis Factor-α Production. Tumor necrosis factor-α (TNF-α) present in the LPS, IL-4, or naïve cell supernatant was determined by commercially available immuno- assay kits (eBioscience Inc., San Diego, CA) and performed as described by the manufacturer. 2.4. Arginase Activity. After 24 h incubation with particles in section 2.2, cells were washed with 500 μL of PBS. In each well, 100 μL of cell lysis buffer (150 μL protease inhibitor cocktail (Amresco, Solon, OH) and 15 μL of Triton X-100 (Acros Organics) filled to 15 mL with DI water) was added, and the plates were incubated on ice for 10 min. Next, 25 μL of the cell lysate was transferred to a 96-well plate, and 25 μL of 10 mM MnCl2 (Fisher) and 50 mM Tris (Fisher) was added to each well. Incubation at 55 °C for 10 min activated the arginase. A solution of 50 μL of 1 M arginine (pH 9.7) was added to each well, and the plate was incubated at 37 °C for 20 h. The activity of arginase was determined by measuring the amount of arginine converted to urea. In 96-well plates, 5 μL of either the urea-containing samples or standards was added to 200 μL of a 1:2 mixture of solution 1 (1.2 g of o-phthaldialdehyde (Alfa Aesar, Ward Hill, MA), 1 L of H2O, and 500 μL of HCl (Fisher, Pittsburgh, PA)) and solution 2 (0.6 g of N-(napthyhl)ethylenediamine dihydrochloride (Acros Organics), 5 g of boric acid (Fisher, Pittsburgh, PA), 800 mL of H2O, and 111 mL of sulfuric acid (Fisher, Pittsburgh, PA), diluted to 1 L with H2O).42 The plates were incubated for 30 min, and the absorbance in each well was measured at 540 nm with a reference at 630 nm using a plate reader. 2.5. Griess Reagent Assay. Nitrite concentration in the supernatant collected in section 2.2 was quantified through a Griess reagent assay. A standard curve was generated through serial dilutions of 100 μM NaNO2. To each well were added 150 μL of sample or standard, 130 μL of DI H2O, and 20 μL of Griess reagent. The plate was incubated for 20 min, and the absorbance of each well was measured with a plate reader at 448 nm with a reference of 690 nm. TNF-α, arginase, and nitrite concentrations were determined at the same time point of 24 h after introduction of the particles. 2.6. Statistics and Data Analysis. Statistical analysis was performed using XLSTAT statistical software (New York, NY). Statistical significance of the mean comparisons was determined by a two-way ANOVA. Pair-wise comparisons were analyzed with Tukey’s honest significant difference test. Differences were considered statistically significant for p < 0.05. The macrophage responses measured above were analyzed by performing partial least-squares (PLS) regression on the material descriptors shown in Table 1. These descriptors were based on descriptors defined by Bicerano.43 3. RESULTS AND DISCUSSION 3.1. Synthesis, ζ-Potential, and Cell Viability. Function- alized p(NIPAm-co-AAc) particles were synthesized and modified with the molecules shown in Figure 1 through the carboxylic acid on the modifier. The ζ-potential of the modified and unmodified p(NIPAm-co-AAc) is in Figure 2. There was a range in ζ-potentials for the particles from −4.2 to −20.8 mV. Cytocompatibility of these materials was tested through an MTT viability assay. Macrophages were activated with LPS or IL-4 to simulate M1- and M2-like cells. After being incubated for 24 h, the medium was replaced and the functionalized particles were added to each well. Metabolic activity of the cells was assessed after an additional 24 h incubation period. Naïve cells, denoted M(0), were also assessed. The results of the viability assay are shown in Figure 3. All tested conditions were >70% viable, with most particles and activation conditions resulting in 90% viability of the macrophages. The largest decrease in viability came from B, D, G, I, and J for M(LPS), H for M(IL-4), and L for M(0). Table 1. Thirty-Eight Molecular Descriptors for the Surface Modifications Used in This Studya ID structure descriptor descriptions 1 R freely rotating bonds 2 HD H-bond donors 3 HA H-bond acceptors 4 Nsp 2 number of sp2 carbon atoms 5 N1°C Number of 1° carbon atoms 6 1 X connectivity index 1, calculated based on the molecular graph of the modifier 7 1 Xv connectivity index 2, calculated based on the electronic configuration of the molecular graph of the modifier 8 0 X atomic index 1, calculated based on the number of non- hydrogen atoms to which a non-hydrogen atom is bound 9 0 Xv atomic index 2, calculated based on the electronic configuration of each non-hydrogen atom 10 1 ξ intensive connectivity index 1, ξ = N 1 X1 11 1 ξv intensive connectivity index 2, ξ = ν ν N 1 X1 12 0 ξ intensive atomic index 1, ξ = N 0 X0 13 0 ξv intensive atomic index 2, ξ = ν ν N 0 X0 14 N number of non-hydrogen atoms 15 NC number of carbon atoms 16 NH number of hydrogen atoms 17 NO number of oxygen atoms 18 NN number of nitrogen atoms 19 NCH2 number of CH2 20 NK NK = 5Namide + 4Nhydroxyl − 3Nether − 5NCC + 3Nsulfone 21 Ndc Ndc = 19NN + 12N(side group O, −S−) + 52Nsulfone − 14Ncyc 22 Ngroup Ngroup = 12Nhydroxyl + 12Namide + 2N(non‑amide −(NH)− unit) − N(alkyl ether −O−) − NCC + 4N(non‑amide −(CO)− next to a nitrogen) + 7N(−(CO)− in carboxylic acid, ketone, or aldehyde) + 2N(other −(CO)−) 23 NMV NMV = −5Nsulfone + 3Nester + 5NCC − 11Ncyc 24 NHη NHη = −7Namide + 6Nether + 5Nsulfone + 3N(six‑membered nonterminal aromatic rings) + 8NCC + 4 N4° carbon atoms + 2N3° carbon atoms 25 NUR NUR = −2NOH − 3Ncyc + 4N(six‑membered aromatic rings) 26 Nds Nds = 2NCH2 − 7NCC + 3N(six‑membered aromatic rings) − 12NCO2 27 NPS NPS = −3Nsp 3 carbon atoms + 6Ncarbonyl groups 28 NvdW NvdW = Nmenonar b + Nalamid c + NOH − 4Ncyc + 2NCC 29 Vw Vw = 3.861803°X + 13.7484351 Xv , van der Waals volume 30 ξNOH ξ = + −N N N NNOH 0.125N O H 31 M molecular weight 32 α polarizability 33 V̅ molar volume 34 log p octanol−water partition ratio 35 log D (pH 5.5) distribution coefficient at pH 5.5 36 log D (pH 7.4) distribution coefficient at pH 7.4 37 ΔHv enthalpy of vaporization 38 γ surface tension a The structure descriptors are defined by Bicerano.43 b Nmenonar is the number of methyl groups attached to nonaromatic atoms. c Nalamid is the total number of linkages between amide and nonaromatic atoms. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 948
  • 4. 3.2. TNF-α Secretion from Activated and Naïve Macrophages. Activated and naïve macrophages were exposed to functionalized p(NIPAm-co-AAc) particles, and the concentration of secreted TNF-α was measured (Figure 4A). LPS was used to stimulate that M1-like phenotype. The control in which no particles were added to the cells contained 4.03 ± 0.51 ng/mL. This was statistically higher (p < 0.05) than the unmodified p(NIPAm-co-AAc) particles and the particles modified with ethylenediamine of 2.49 ± 0.29 and 2.55 ± 0.25 ng/mL, respectively. Compared to both the unmodified and Eda-modified particles, modifications D and I−M resulted in statistically higher TNF-α secretion (p < 0.05). Of these, albizziin increased TNF-α secretion ∼2-fold. Modifications B, E, G, and H decreased the TNF-α production (p < 0.05). The most significant decrease was observed for 3-guanidinopro- pionic acid of ∼40%. Details of multiple comparisons for these data are in Table S1. To explore the M2-like phenotype, macrophages were stimulated with IL-4. The control without particles resulted in 2.69 ± 0.08 ng/mL TNF-α measured in the supernatant. The unmodified p(NIPAm-co-AAc) and Eda-modified particles resulted in a decrease in TNF-α by 1.78 ± 0.13 and 1.68 ± 0.12 ng/mL, respectively (p < 0.05), similar to the M(LPS) cells. Modifications A, I−K, and N resulted in increased TNF-α secretion (p < 0.05), while modifications B, C, E, F, and H resulted in a decrease (p < 0.05) compared to the unmodified and Eda particles. Modifications C−G and K resulted in increases in TNF-α secretion, and particles modified with I, J, L, and M decreased the TNF-α concentration detected compared to the unmodified and Eda particles. 2-Amino-3- guanidinopropionic caused the largest increase in TNF-α secretion of ∼200%, and 3-guanidinopropionic acid caused the most significant decrease of ∼40%. Details of multiple comparisons for these data are in Table S2. Macrophages without activation were also incubated with the modified particles, and the secreted TNF-α was assessed. The control, unmodified p(NIPAm-co-AAc), and Eda particles were Figure 2. ζ-Potentials for modified p(NIPAm-co-AAc) particles. Data represent the mean value of three replicates for each sample ± standard deviation. Eda = ethylenediamine, pN = unmodified p(NIPAm-co-AAc). Figure 3. Viability assay showing the cytotoxicity of all particles with LPS or IL-4 activated or naïve RAW 264.7 cells. The % viability was normalized to RAW 264.7 cells treated with LPS, IL-4, or naïve cells without particles. Data represent the mean value of four replicates for each sample ± standard deviation. Eda = ethylenediamine, pN = unmodified p(NIPAm-co-AAc). Figure 4. Molecular expression of (A) TNF-α and (B) urea/nitrite by macrophages in respose to functionalized p(NIPAm-co-AAc) particles and stimulation with LPS or IL-4. Naïve cells are also shown. Data represent the mean value of four replicates for each sample ± standard deviation. Eda = ethylenediamine, pN = unmodified p(NIPAm-co- AAc), PC = positive control of cells without materials. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 949
  • 5. not statistically different with 1.04 ± 0.11, 1.31 ± 0.11, and 1.12 ± 0.13 ng/mL TNF-α detected. The largest increase in TNF-α was found for modifications F and K, with an increase of ∼350%. The largest decrease was observed for modifications I, J, and L, resulting in an 80% decrease in TNF-α compared with the unmodified or Eda particles. Details of multiple comparisons are in Table S3. Taken together, modifications I, J, and K lead to increases in TNF-α for both M(LPS) and M(IL-4), while modifications B, E, and H lead to decreases in TNF-α secretion. The most dramatic decreases in TNF-α secretion for both M(LPS) and M(IL-4) were in the presence of the 3-guanidinopropionic acid modification, which reduced TNF-α secretion by ∼40% compared to unmodified and Eda-modified particles. 3.3. Urea/Nitrite for Activated and Naïve Macro- phages. Arginase activity was measured by exposing cell lysate to arginine and assessing its conversion to urea after 20 h incubation. Increased arginase activity is suggestive of an M2- like phenotype. Increased iNOS indicates an M1-like phenotype. The activity of iNOS was indirectly measured through a Griess assay in which nitrites, the stable form of NO, are quantified. NO results from reactive nitrogen intermediates, which are synthesized by iNOS. Arginine is generally metabolized through one of these pathways, thus the ratio of urea/nitrite can assess the extent of polarization toward either phenotype.25,26 These results are shown in Figure 4B. The nitrite and urea concentrations are shown as Figure S1A,B. M(IL-4) cells were found to have 447 ± 68 mg/μmol urea/ nitrite. Cells incubated with A−D and I−L increased the amount of urea/nitrite measured (p < 0.05), while particles modified with E−H decreased the levels of urea/nitrite (p < 0.05). Measured levels of urea/nitrite ranged from 252 to 1023 mg/μmol. There was no statistical difference between the M(IL-4) cells in the absence of particles and M(LPS) cells incubated with unmodified and Eda-modified p(NIPAm-co- AAc) particles: 421 ± 100 and 366 ± 70 mg/μmol urea/nitrite, respectively. Details of multiple comparisons for urea/nitrite for M(IL-4) are in Table S4. M1-like cells were also studied using M(LPS)-stimulated cells. No statistical differences were observed between the control and cells incubated with unmodified and Eda-modified particles: 62 ± 5, 69 ± 6, and 70 ± 7 mg/μmol urea/nitrite, respectively. Particles E−H, K, and M resulted in elevated urea/ nitrite levels. Only particles A and I decreased urea/nitrite (p < 0.05) compared to the positive control, unmodified, and Eda- modified particles. Details of multiple comparisons for urea/ nitrite for M(LPS) are provided in Table S5. In comparing trends between M(LPS) and M(IL-4), only modification K resulted in increasing urea/nitrite levels and no modifications were able to decrease urea/nitirite in both M(LPS) and M(IL- 4) cells. It is important to note that these results may be particular to murine macrophage cells. Human macrophages transport arginine through system y+ L transporters,44 whereas a cationic amino acid transporter has been suggested as an arginine transporter in mice.45 Naïve macrophages were also tested for urea/nitrite levels. The levels of nitrites detected for all M(0) cells were lower than the standard curve. Statistical analysis on these values is not reasonable. 3.4. Material Parameters That Influence Macrophage Phenotypic Shifts. Secretion of TNF-α in the absence of particles followed the expected trend, in which the highest levels were observed for M(LPS), followed by M(IL-4) and M(0).6,46−48 With the exception of modification A, M(LPS) cells secreted more TNF-α than the M(IL-4) cells when they were incubated with particles. The M(0) cells exhibited a range of responses to the particles, secreting less TNF-α in the presence of modifications A, C, J, Eda, and unmodified p(NIPAm-co-AAc) than M(LPS) cells. Increased TNF-α was observed for modification M compared to M(LPS) cells. The increase in TNF-α secretion may be a result of LPS tolerance developed in the M(LPS) cells, which leads to a decreased production of TNF-α.49 Another possibility is that IL-10, a suppressor of TNF-α, may be secreted. Previous findings have demonstrated a decrease of TNF-α secretion in M(LPS) cells by a factor of 21.4 ± 2.5 in the presence of 0.1 ng/mL IL-10.50 Arginase/iNOS is a measure of macrophage polarization,25,26 with high levels denoting M2-like states and low levels indicating a more M1-like state. Here, we measure this using urea/nitrite levels. In the case of M(IL-4), both the control and the cells incubated with particles resulted in higher urea/nitrite levels, which is expected. The aim of this work was to identify material properties that influence M1- and M2-like phenotypes. The trends between material properties and cell responses were quantitatively modeled using material descriptors listed in Table 1. These descriptors were based largely on structural descriptors43 along with physiochemical properties. PLS was performed on the measured data using principle component analysis values of Figure 4A,B as a function of the molecular descriptors in Table 1. The results of the model are presented in Figure 5, where the horizontal axis represents TNF-α secretion and the vertical axis is urea/nitrite. Cells that secrete high levels of TNF-α and exhibit low levels of urea/nitrite are M1-like, while the converse are M2-like. Both of these regions are outlined in Figure 5. In addition, the specific descriptors that promote either of these two extremes are labeled on the plots. For M(IL-4) cells (Figure 5A), increasing H-bond donors promoted a shift toward an M1-like phenotype, while increasing H-bond acceptors, Ndc, log D (5.5), and log D (7.4) retained the M2- like phenotype. Log D is the octanol−water distribution coefficient, which can be measured at any pH, and Ndc is a correction factor for the dielectric constant. It bears mentioning that the inverse of these material descriptors would result in a shift toward the opposite phenotype. Based on these parameters, polarity of the molecule appears to be an important factor in altering M(IL-4) polarization. It should be noted that surface tension had a negligible effect on either TNF-α or urea/ nitrite. A larger number of material properties were identified as altering M(LPS), shown in Figure 5B. Increasing the number of freely rotating bonds, aliphatic carbons, van der Waals interactions (calculated using NνdW), the number of carbon and hydrogen atoms, and the enthalpy of vaporization promotes an M2-like phenotype. On the other end of the spectrum, primary carbon atoms, the number of sp2 carbon atoms, NMVa correction term for molar volume, NURa correction term for molar Rao function, ξNOHa correction term for thermal conductivity, log p, log D (7.4), and log D (5.5) promoted the M1-like phenotype. All of these parameters suggest that hydrophobicity plays a dominant role in altering M(LPS) cells. Since the levels of nitrites detected for all M(0) cells were lower than the standard curve, material properties influencing phenotypic shifts were not analyzed because statistical analysis on these values is not reasonable. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 950
  • 6. Interesting similarities exist for the material properties that promote phenotypic shifts in all the conditions examined here. Descriptors related to hydrophobicity, van der Waals interactions, and H-bonding were found to be related to macrophage polarization for M(LPS) and M(IL-4). The conventional dogma surrounding ligand−receptor binding involves all of these interaction forces additively combining to stabilize the ligand−receptor pair.51 Very likely, serum proteins present in the culture media adsorb in different configurations on the particle surface, thus enabling different interactions with surface receptors. The complexity of macrophage activation and reprogramming through surface receptors prevents predicting which receptors these particles are interacting with; however, examining the impact of material descriptors on altering macrophage response has the potential to improve material systems for implanted biomaterials as well as drug delivery systems. Based on the observations described above and in Figure 5, three molecules were selected to alter the macrophage phenotype: lysine, ornithine, and ureidopropionic acid. These molecules were conjugated to Eda-modified particles as described in section 2.1.1. The concentration of secreted TNF-α and urea/nitrite in response to incubation with the modified particles is shown in Figure 6. Ureidopropionic acid was predicted to promote urea/nitrite and TNF-α secretion in M(IL-4) cells and decrease secretion in M(LPS). Factors that would increase urea/nitrite and TNF-α secretion in M(IL-4) cells would be increased distribution coefficients at pH 5.5 and 7 and a reduction in H-bond acceptors. Compared to albizziin, ureidopropionic acid has fewer H-bond acceptors and higher distribution coefficients at pH 5.5 (−4.0 vs −2.62, respectively). The distribution coefficients at pH 7 are decreased for ureidopropionic acid (−4.4 vs −4.01). The distribution coefficients at pH 5.5 and 7 are very similar when compared to those of albizziin to citrulline, ergo an increase in urea/nitrite and TNF-α secretion would be expected for ureidopropionic acid compared to citrulline, as is observed in Figure 5A,B, particularly since citrulline has an additional H-bond donor compared to ureidoprionic acid, much like albizziin. In the case of M(LPS) cells, factors that cause cells to secrete decreased levels of urea/ nitrite are the distribution coefficients at pH 5.5 and 7. In the Figure 5. Analysis of molecular descriptors to identify polymer characteristics that promote M1- or M2-like profiles. Pearson’s correlation between molecular descriptors that promote urea/nitrite (y-axis) and TNF-α (x-axis) for (A) IL-4 and (B) LPS. Relevant descriptors that promote M1- or M2-like responses are labeled on the figure. Figure 6. Molecular expression of (A) TNF-α and (B) urea/nitrite by macrophages in respose to functionalized p(NIPAm-co-AAc) particles and stimulation with LPS or IL-4. Naïve cells are also shown. Data represent the mean value of four replicates for each sample ± standard deviation. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 951
  • 7. case of M(LPS) cells, the higher distribution coefficient for ureidopropionic acid compared to that of either albizziin or citrulline would cause a decrease in the urea/nitrite and TNF-α levels, as was observed. The number of freely rotating bonds is also lower for ureidopropionic acid compared to albizziin and citrulline. The difference between lysine and ornithine is an additional aliphatic CH2 group in lysine, which also results in an additional freely rotating bond for lysine. The other parameters influencing urea/nitrite production are similar for the two modifiers. This resulted in predictions that lysine would result in increased nitrite/urea secretion compared to ornithine. The additional CH2 group results in a 2-fold increase in urea/nitrite for the two modifiers in the case of M(LPS) cells. In examining the response of M(IL-4) cells, no difference between lysine- and ornithine-modified particles was expected for urea/nitrite and TNF-α levels since the differences in their structures did not result in differences in the material descriptors identified in Figure 5A,B. 4. CONCLUSIONS In summary, we have examined how a library of 14 surface modifiers alters macrophage response and have correlated those responses to material descriptors that promote shifts in phenotypes. Similarities between all three macrophage treat- ment conditions exist in that intermolecular nonbonding interactions play an important role in altering macrophage responses. The differences between these three conditions suggest the possibility that material properties can be exploited to manipulate macrophage responses, possibly reprogramming these cells without exogenous protein delivery. Semiquantita- tive methods such as those employed in this study have the potential to both improve screening for materials that promote M1- or M2-like phenotypes in macrophages and add to the current library of knowledge pertaining to how materials influence cellular responses. ■ ASSOCIATED CONTENT *S Supporting Information The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsbiomater- ials.6b00041. Details of multiple comparisons for TNF-α for M(LPS), M(IL-4), and M(0) cells and for urea/nitrite for M(LPS) and M(IL-4) cells; nitrite and urea measured for M(LPS), M(IL-4), and M(0) cells (PDF) ■ AUTHOR INFORMATION Corresponding Author *Tel: 515-294-7304. Fax: 515-294-5444. E-mail: kbratlie@ iastate.edu. Notes The authors declare no competing financial interest. ■ ACKNOWLEDGMENTS This work was supported by the National Science Foundation under Grant No. CBET 1227867 and the Roy J. Carver Charitable Trust Grant No. 13-4265. The authors also acknowledge support from NSF ARI-R2 (CMMI-0963224) for funding the renovation of the research laboratories used for these studies. ■ REFERENCES (1) Acharya, A. P.; Dolgova, N. V.; Clare-Salzler, M. J.; Keselowsky, B. G. Adhesive substrate-modulation of adaptive immune responses. Biomaterials 2008, 29, 4736−50. (2) Schutte, R. J.; Xie, L.; Klitzman, B.; Reichert, W. M. In vivo cytokine-associated responses to biomaterials. Biomaterials 2009, 30, 160−8. (3) Anderson, J. M.; Rodriguez, A.; Chang, D. T. Foreign body reaction to biomaterials. Semin. Immunol. 2008, 20, 86−100. (4) Nilsson, B.; Ekdahl, K. N.; Mollnes, T. E.; Lambris, J. D. The role of complement in biomaterial-induced inflammation. Mol. Immunol. 2007, 44, 82−94. (5) Wang, D.; Phan, N.; Isely, C.; Bruene, L.; Bratlie, K. M. The effect of polymer chemistry and macrophage phenotype on particle internalization. Biomacromolecules 2014, 15, 4102−10. (6) Wang, D.; Bratlie, K. M. Influence of Polymer Chemistry on Cytokine Secretion from Polarized Macrophages. ACS Biomater. Sci. Eng. 2015, 1, 166−74. (7) Bygd, H. C.; Akilbekova, D.; Muñoz, A.; Forsmark, K. D.; Bratlie, K. M. Poly-l-arginine based materials as instructive substrates for fibroblast synthesis of collagen. Biomaterials 2015, 63, 47−57. (8) Bygd, H. C.; Forsmark, K. D.; Bratlie, K. M. Altering in vivo macrophage responses with modified polymer properties. Biomaterials 2015, 56, 187−97. (9) Petersen, L. K.; Xue, L.; Wannemuehler, M. J.; Rajan, K.; Narasimhan, B. The simultaneous effect of polymer chemistry and device geometry on the in vitro activation of murine dendritic cells. Biomaterials 2009, 30, 5131−42. (10) Ariganello, M. B.; Simionescu, D. T.; Labow, R. S.; Lee, J. M. Macrophage differentiation and polarization on a decellularized pericardial biomaterial. Biomaterials 2011, 32, 439−49. (11) Rodriguez, A.; Meyerson, H.; Anderson, J. M. Quantitative in vivo cytokine analysis at synthetic biomaterial implant sites. J. Biomed. Mater. Res., Part A 2008, 89, 152−9. (12) Mosser, D. M. The many faces of macrophage activation. J. Leukocyte Biol. 2003, 73, 209−12. (13) Murray, P. J.; Allen, J. E.; Biswas, S. K.; Fisher, E. A.; Gilroy, D. W.; Goerdt, S.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14− 20. (14) Mantovani, A.; Locati, M. Tumor-associated macrophages as a paradigm of macrophage plasticity, diversity, and polarization: lessons and open questions. Arterioscler., Thromb., Vasc. Biol. 2013, 33, 1478− 83. (15) Biswas, S. K.; Sica, A.; Lewis, C. E. Plasticity of Macrophage Function during Tumor Progression: Regulation by Distinct Molecular Mechanisms. J. Immunol. 2008, 180, 2011−7. (16) Qian, B.-Z.; Pollard, J. W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39−51. (17) Gollob, J A.; Mier, J. W.; Veenstra, K.; McDermott, D. F.; Clancy, D.; Clancy, M.; et al. Phase I trial of twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant melanoma: ability to maintain IFN-gamma induction is associated with clinical response. Clin. Cancer Res. 2000, 6, 1678−92. (18) Hill, H.; Conway, T.; Sabel, M.; Jong, Y.; Mathiowitz, E.; Bankert, R.; et al. Cancer immunotherapy with interleukin 12 and granulocyte-macrophage colony-stimulating factor-encapsulated mi- crospheres: coinduction of innate and adaptive antitumor immunity and cure of disseminated disease. Cancer Res. 2002, 62, 7254−63. (19) Watkins, S. K.; Egilmez, N. K.; Suttles, J.; Stout, R. D. IL-12 rapidly alters the functional profile of tumor-associated and tumor- inflitrating macrophages in vitro and in vivo. J. Immunol. 2007, 178, 1357−62. (20) Hofkens, W.; Schelbergen, R.; Storm, G.; van den Berg, W. B.; van Lent, P. L. Liposomal Targeting of Prednisolone Phosphate to Synovial Lining Macrophages during Experimental Arthritis Inhibits M1 Activation but Does Not Favor M2 Differentiation. PLoS One 2013, 8, e54016. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 952
  • 8. (21) Jain, S.; Tran, T.-H.; Amiji, M. Macrophage repolarization with targeted alginate nanoparticles containing IL-10 plasmid DNA for the treatment of experimental arthritis. Biomaterials 2015, 61, 162−77. (22) Bronte, V.; Zanovello, P. Regulation of immune responses by L- arginine metabolism. Nat. Rev. Immunol. 2005, 5, 641−54. (23) Morris, S. M. Arginine metabolism: boundaries of our knowledge. J. Nutr. 2007, 137, 1602S−1609S. (24) Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677−86. (25) Lumeng, C. N.; Bodzin, J. L.; Saltiel, A. R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 2007, 117, 175−84. (26) Allen, J. E.; Loke, P. Divergent roles for macrophages in lymphatic filariasis. Parasite Immunol. 2001, 23, 345−52. (27) Murphy, B. S.; Sundareshan, V.; Cory, T. J.; Hayes, D.; Anstead, M. I.; Feola, D. J. Azithromycin alters macrophage phenotype. J. Antimicrob. Chemother. 2008, 61, 554−60. (28) Rath, M.; Muller, I.; Kropf, P.; Closs, E. I.; Munder, M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 1−10. (29) Amat di San Filippo, C.; Taylor, M. R. G.; Mestroni, L.; Botto, L. D.; Longo, N. Cardiomyopathy and carnitine deficiency. Mol. Genet. Metab. 2008, 94, 162−6. (30) Ferrante, R.; Andreassen, O.; Jenkins, B.; Dedeoglu, a; Kuemmerle, S.; Kubilus, J.; et al. Neuroprotective effects of creatine in a transgenic mouse model of Huntington’s disease. J. Neurosci. 2000, 20, 4389−97. (31) GUALANO, B.; DE SALLES PAINNELI, V.; ROSCHEL, H.; ARTIOLI, G. G.; NEVES, M.; DE SÁ PINTO, A. L.; et al. Creatine in Type 2 Diabetes. Med. Sci. Sports Exercise 2011, 43, 770−8. (32) Arimoto, T.; Bing, G. Up-regulation of inducible nitric oxide synthase in the substantia nigra by lipopolysaccharide causes microglial activation and neurodegeneration. Neurobiol. Dis. 2003, 12, 35−45. (33) Grant, S. K.; Green, B. G.; Stiffey-Wilusz, J.; Durette, P. L.; Shah, S. K.; Kozarich, J. W. Structural requirements for human inducible nitric oxide synthase substrates and substrate analogue inhibitors. Biochemistry 1998, 37, 4174−80. (34) Jang, J. J.; Ho, H. K.; Kwan, H. H.; Fajardo, L. F.; Cooke, J. P. Angiogenesis is impaired by hypercholesterolemia: role of asymmetric dimethylarginine. Circulation 2000, 102, 1414−9. (35) Koc, a; Ozkan, T.; Karabay, a Z; Sunguroglu, a; Aktan, F. Effect of L-carnitine on the synthesis of nitric oxide in RAW 264·7 murine macrophage cell line. Cell Biochem Funct 2011, 29, 679−85. (36) Oudman, I.; Clark, J. F.; Brewster, L. M. The Effect of the Creatine Analogue Beta-guanidinopropionic Acid on Energy Metab- olism: A Systematic Review. PLoS One 2013, 8, e52879. (37) Hagen, T. M.; Ingersoll, R. T.; Wehr, C. M.; Lykkesfeldt, J.; Vinarsky, V.; Bartholomew, J. C.; et al. Acetyl-L-carnitine fed to old rats partially restores mitochondrial function and ambulatory activity. Proc. Natl. Acad. Sci. U. S. A. 1998, 95, 9562−6. (38) Calabrese, V.; Scapagnini, G.; Ravagna, a.; Bella, R.; Butterfield, D a.; Calvani, M.; et al. Disruption of Thiol Homeostasis and Nitrosative Stress in the Cerebrospinal Fluid of Patients with Active Multiple Sclerosis: Evidence for a Protective Role of Acetylcarnitine. Neurochem. Res. 2003, 28, 1321−8. (39) Ricote, M.; Li, A. C.; Willson, T. M.; Kelly, C. J.; Glass, C. K. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 1998, 391, 79−82. (40) Guastadisegni, C.; Minghetti, L.; Nicolini, A.; Polazzi, E.; Ade, P.; Balduzzi, M.; et al. Prostaglandin E2 synthesis is differentially affected by reactive nitrogen intermediates in cultured rat microglia and RAW 264.7 cells. FEBS Lett. 1997, 413, 314−8. (41) Motrán, C. C.; Díaz, F. L.; Gruppi, A.; Slavin, D.; Chatton, B.; Bocco, J. L. Human pregnancy-specific glycoprotein 1a (PSG1a) induces alternative activation in human and mouse monocytes and suppresses the accessory cell-dependent T cell proliferation. J. Leukocyte Biol. 2002, 72, 512−21. (42) Levinson, S. S. Kinetic centrifugal analyzer and manual determination of serum urea nitrogen, with use of o-phthaldialdehyde reagent. Clin. Chem. 1978, 24, 2199−202. (43) Bicerano, J. Prediction of Polymer Properties; Marcel Dekker Inc.: New York, 2002. (44) Rath, M.; Muller, I.; Kropf, P.; Closs, E. I.; Munder, M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front. Immunol. 2014, 5, 1−10. (45) Manner, C. K.; Nicholson, B.; MacLeod, C. L. CAT2 arginine transporter deficiency significantly reduces iNOS-mediated NO production in astrocytes. J. Neurochem. 2003, 85, 476−82. (46) Akilbekova, D.; Philiph, R.; Graham, A.; Bratlie, K. M. Macrophage reprogramming: influence of latex beads with various functional groups on macrophage phenotype and phagocytic uptake in vitro. J. Biomed. Mater. Res., Part A 2015, 103, 262. (47) Barth, K a.; Waterfield, J. D.; Brunette, D. M. The effect of surface roughness on RAW 264.7 macrophage phenotype. J. Biomed. Mater. Res., Part A 2013, 101, 2679−88. (48) Varin, A.; Mukhopadhyay, S.; Herbein, G.; Gordon, S. Alternative activation of macrophages by IL-4 impairs phagocytosis of pathogens but potentiates microbial-induced signalling and cytokine secretion. Blood 2010, 115, 353−62. (49) Docke, F. R. W.; Bundschuhrt, D. S.; Hartung, T.; Wendel, A.; Volk, H. In vitro prevention and reversal of lipopolysaccharide desensitization by IFN-gamma, IL-12, and granulocyte-macrophage colony-stimulating factor. J. Immunol. 1997, 158, 2911−8. (50) Bogdan, C.; Vodovotz, Y.; Nathan, C. Macrophage deactivation by interleukin 10. J. Exp. Med. 1991, 174, 1549−55. (51) Seong, S.-Y.; Matzinger, P. Hydrophobicity: an ancient damage- associated molecular pattern that initiates innate immune responses. Nat. Rev. Immunol. 2004, 4, 469−78. ACS Biomaterials Science & Engineering Article DOI: 10.1021/acsbiomaterials.6b00041 ACS Biomater. Sci. Eng. 2016, 2, 946−953 953