Final Thesis (1) - Merged - Compressed
Final Thesis (1) - Merged - Compressed
Final Thesis (1) - Merged - Compressed
II
Dedication
((Iraq))
To My daughter, Asawer,
To My son, Yaman,
Ghassan
III
Acknowledgments
It is of great pleasure for me to express my sincere
appreciation to my supervisor Dr. Tarek Rebai, Professor in
Laboratory Histo-embryology and Cytogenetics department,
Faculty of Medicine of Sfax, Tunisia, for his supervision,
guidance, understanding, patience, motivation, and assistance to
start and successfully finish this work.
My thanks go to Prof. Dr. Ali Mohammed Ghazi Vet.
Medicine college of Al-Qadisiyah, Iraq, for his effort in the
statistical analysis. Special thanks to Assistant prof. Dr. Hassan
Khalaf Ulaiwi Al-Karagoly in Vet. Medicine college of Al-
Qadisiyah, Iraq, for helping in the reading and analysis of the
histopathological sections. I would like to express sincere thanks
to instructor Dr. Abdulrazzaq B. Kadhim in Vet. Medicine
College of Al-Qadisiyah, Iraq, for helping me Perform the
laboratory animal anatomy. I would like to thank for Mrs.
Mounira Hmani-Aifa (Committee President), Mrs. Noura
Bougacha (Reporter), Mrs. Rim Frikha (Reporter), Mr. Mongi
Saoudi (Examiner) for their valuable advices and sound
guidance in evaluating this work.
I would also like to thank everyone who supported me and
helped me, and provided the appropriate environment for the
success of this current research plan, even with a kind word.
Ghassan
IV
The content list
Title N.
- Title
- List Of Publications II
- Dedication III
- Acknowledgments IV
- Abbreviations List XII
Abstract XIV
- Introduction 1
- Aims of the study 3
1. Literature 4
1.1. The stem cell history 4
1.2 Classification of the stem cells 5
I Types of stem cells according to the source 5
II Types of stem cells according to the differentiation 16
1.3. Therapeutic uses of the stem cells 20
1.4. Lupus disease 22
1.4.1. Lupus Epidemiology 23
1.4.2. Causes of lupus 25
1.4.3. Types of LUPUS 25
1.4.4. Clinical signs of lupus 26
1.4.5. Pathophysiology of the lupus 27
1.4.6. The genes related to the Lupus 32
1.4.7. Lupus Diagnosis 32
1.4.8. Lupus treatment 33
1.5. Role of the mesenchymal stem cells in lupus therapy 34
Materials and Methods 36
1.1 The materials 37
1.2. The study animals 38
1.3. The study designs 39
1.4. BALB/C model mice 39
1.5. The experimental designs 40
1.6. Collection of blood 42
1.7. The lupus diagnosis confirmation 43
1.8. The BALB/c BM-MSCs preparation 43
1.9. Administration of BALB/c- BM- MSCs 44
V
1.10. The used ELISA kits 44
1.10.1. Mouse Anti-nuclear antibody (ANA) ELISA Kit 44
1.10.2. Mouse anti -ds DNA ELISA Kit 45
1.10.3. Mouse CCL2 (MCP-1) Uncoated ELISA 45
1.10.4. Mouse RANTES ELISA Kit (CCL5) 45
1.10.5 Mouse ICAM-1 ELISA Kit 46
1.10.6. Mouse IL‑6 ELISA Kit 46
1.10.7. Mouse IL-10 ELISA Kit 46
1.10.8. Mouse Interferon gamma ELISA Kit 47
1.10.9 Mouse TGF beta 1 ELISA Kit 47
1.10.10 Mouse VEGF kit 48
1.11 The molecular examination 48
1.11.1 The used primers 49
1.11.2 Total RNA Extraction and Reverse Transcriptase 66
1.12. Histopathological examination 49
1.13. Morphometry 51
1.14. Statistical analysis 51
The results and the discussion 52
1.1. ANA and Anti dsDNA levels (The serological tests) 52
1.2. The cytokine and chemokines levels (Serological tests) 54
1.3. The molecular examination (RT-PCR) 60
1.4. The histopathological examination 70
1.4.1 Liver 70
1.4.2 Kidney 75
1.5. The correlation study 81
The conclusion and Perspectives 87
1.1. The conclusion 87
1.2. The Perspectives 89
References 91
Annexes 118
The published papers 153
VI
The tables list
VII
The figures list
IX
of hepatocytes (blue arrow), and dilation of sinusoids
(black arrow) (G1). H&E, 400X.
Figure 32 A- Normal liver architecture. H&E, 40X. B- Normal 71
hepatocytes (black arrows), focal area of necrosis (blue
arrow). H&E, 100X. C- The normal hepatocytes with
prominent nuclei and deeply eosinophilic cytoplasm;
necrotic hepatocytes and sinusoids dilation (G2). H&E,
400X.
Figure 33 Liver Section with normal liver architectures. H&E 72
(G3), 40X.
Figure 34 Liver Section with normal liver architectures (G3). 72
H&E, 100X.
Figure 35 Liver section with normal hepatocytes. H&E, 400X 72
(control group)
Figure 36 Kidney section with atrophy of glomeruli (black arrow); 75
some glomeruli are normal (yellow arrows) (G1), H&E,
4X and 400X.
Figure 37 The severe necrosis of renal tubules of the kidney (blue 76
arrows), vascular dilatation (yellow arrow), obliteration
of some renal tubules (black arrow), fibrosis (pink
arrow) of interstitial tissue between renal tubules,
hemorrhage (H), and deposition of eosinophilic
materials (green arrow) in the lumen of the renal tubule
(G1) H&E, 400X.
Figure 38 Almost normal renal tubules of the kidney, but 76
sloughing of the epithelial lining of a few renal tubules
(yellow arrows) was also seen (G2), H&E, 400X.
Figure 39 Normal renal glomeruli of the kidney (black arrows) 77
and few atrophied glomeruli (blue arrow) were also
seen; almost normal renal tubules with slightly sloughed
lining epithelium (yellow arrows) of renal tubules were
evident (G2). H&E, 40X and 400X.
Figure 40 Normal renal glomeruli of the kidney (black arrows) 77
and renal tubules, but mild sloughing of lining
epithelium (red arrow) were also evident (G2). H&E,
40X, 100X, and 400X.
Figure 41 Normal renal glomeruli of the kidney (black arrows) 78
and renal tubules in (G3). H&E, 400X.
Figure 42 Normal renal glomeruli and renal tubules of the kidney 78
X
(G4) (control group), H&E, 100X and 400X
XI
Abbreviations List
XII
(MSC-EVs) MSC-derived extracellular vesicles
(MSCs) Multipotent mesenchymal stem cells
(MSCT) MSC transplantation
(NSC) Nullipotent stem cells
(NZB/NZW) hybrid New Zealand mice
(OS) Overlap Syndromes
(OSC) Oligopotent stem cells
(PB) Peripheral blood
(PBS) Phosphate-buffered saline
(PGCs) Primordial germ cells
(PSC) Pluripotent stem cells
(SCLE) Subacute Cutaneous Lupus Erythematosus
(SCNT) Somatic cell nuclear transfer
(SLE) Systemic Lupus Erythematosus
(SLEDAI) SLE disease activity index
(ST-HSC) Short term HSC
(Tfh) T follicular helper
(Tfh) Th1, Th2, T follicular helper
(TGF-β1) Transforming growth factor beta1
(Th) T helper
(TSC) Totipotent stem cells
(U.S.FDA) United States food and agriculture association
(UCB) Umbilical cord blood
(UC-MSCc) Umbilical cord mesenchymal stem cell
(USC) Unipotent stem cells
(WJ-MSCs) Wharton's jelly mesenchymal stem cells
XIII
Abstract:
Systemic lupus erythematosus (SLE) causes inflammatory reactions in the
body's organs. SLE causes disturbance and bad regulation of the immune response,
particularly in the immune cells and cytokines. The work aims to evaluate the
levels of some cytokines levels in mice with SLE, and then treated by bone marrow
Mesenchymal stem cells (BM-MSCs), determine the expression genes of some of
the cytokines encoding genes, and investigate the histopathological changes in the
liver and kidney. This study was carried out for the first time, especially
concerning studying most of the cytokines that have a close relationship with lupus
disease and also with regard to studying the genes that encode those cytokines to
determine the relationship between them. (40) Male BALB/c mice are randomly
divided into four groups, each consisting of ten mice. The experiment study was
done in the animal house of Veterinary Medicine College, Al-Qadisiyah
University. G1 is injected activated lymphocytes Derivative (ALD-DNA) S/C
(three doses) at the (0, 14, and 28) day of the experiment beginning for induction
lupus. G2 was injected (ALD-DNA) S/C (three doses) on the (0, 14, and 28) day of
the experiment beginning for induction SLE, then treated with a single dose of
BM-MSCs (0.5×106) IV for 10 grams. G3 was administered a single dose of BM-
MSCs (0.5×106) IV for 10 grams. G4 administrated PBS only. After finishing the
induction and treatment time, all the animals were sacrificed to collect the blood
and tissue samples to determine the cytokines and chemokines levels by Eliza kits
and determine the gene expression of the cytokines and chemokines encoding
genes by RT-PCR techniques; moreover, determine the histopathological changes
of the liver and kidney. The results showed a significant increase in ANA and anti-
dsDNA in G1 and a decrease in G2 compared with G3 and G4. There is no
significant difference between G3 and G4 in ANA and anti-dsDNA levels. G1
showed a significant increase in IL-6, CCL-5/RANTES, VEGF, ICAM, CCL-2,
and IFNγ levels and a decrease in IL-10 and TGFβ1 than in the control group. G2
reveals low levels of IL-6, CCL-5/RANTES, VEGF, ICAM, CCL-2/MCP-1, and
IFNγ but showed higher levels of IL-10 and TGFβ1 than in control. Furthermore,
G3 has no significant differences from G4 at. G1 showed significantly higher gene
expression of IL-6, CCL-5/RANTES, VEGF, ICAM, CCL-2, and IFNγ-encoding
genes and low gene expression of IL-10 and TGFβ1-encoding genes than in
control. G2 reveals a low significant gene expression of IL-6, CCL-5/RANTES,
VEGF, ICAM, CCL-2/MCP-1, and IFNγ-encoding genes while showing
significantly high gene expression of IL-10 and TGFβ1-encoding gene as
compared with the control group. G3 doesn't show any changes in the gene
expression of the cytokines and chemokines as compared to the control group. The
histopathological changes in the liver in G1 included hepatocyte necrosis, dilated
sinusoids, hemorrhage, and normal striation of hepatocyte loss. G2 showed normal
XIV
hepatocytes but also a focal area of necrosis. G3 and G4 showed normal
architecture of the liver tissue. The kidney in G1 showed glomeruli atrophy, tubule
necrosis, vascular dilatation, obliteration of some tubules, and interstitial fibrosis.
There are hemorrhages and deposition of eosinophilia in the tubule lumen. G2
showed almost normal tubules but slight sloughing of the epithelial lining of a few
tubules and a few atrophied glomeruli. G3 and G4 have normal glomeruli, normal
proximal, distal, and convoluted tubules. The correlation study was done between
the used cytokines and chemokines and showed that G1 showed a positive
correlation between IL-10 and IL-6 and CCL2 and VEGF. However, it showed a
negative correlation between IL-10 and CCL2, IL-10 and VEGF, IL-6 and VEGF,
and TGF- β1 and CCL2. G2 doesn't show any correlation relationship between the
used parameters. G3 showed a negative correlation between TGF β1 and CCL-2
and IL-6 and INF- γ, while the rest of the relationships between the criteria used
did not show a clear correlation relationship. G4 doesn't show a relationship
between the used parameters, except correlation between CCL-5 and INF- γ
(positive correlation) and between TGF β1 and VEGF (negative correlation). BM-
MSCs have an essential and effective therapeutic role in treating lupus at the
serological and molecular levels of the cytokines and the histopathological levels
by regulating cytokine production and gene expression of the cytokines treating the
lupus disease.
Keywords: ELIZA, RT-PCR, mice, mesenchymal stem cells, cytokines, Balb/c
SLE
XV
Résumé:
XVI
génique significative de l'IL-6, du CCL-5/RANTES, du VEGF, de l'ICAM, du
CCL-2/MCP-1 et des gènes codant pour l'IFNγ, tout en montrant une expression
génique significativement élevée des gènes codant pour l'IL-10 et le TGFβ1 par
rapport au groupe témoin. G3 ne montre aucun changement dans l'expression
génique des cytokines et des chimiokines par rapport au groupe témoin. Les
changements histopathologiques dans le foie de G1 comprenaient une nécrose
hépatocytaire, des sinusoïdes dilatés, des hémorragies et une perte normale de
striation des hépatocytes. G2 a montré des hépatocytes normaux mais aussi une
zone focale de nécrose. G3 et G4 ont montré une architecture normale du tissu
hépatique. Le rein de G1 a montré une atrophie des glomérules, une nécrose des
tubules, une dilatation vasculaire, une oblitération de certains tubules et une fibrose
interstitielle. Il y a des hémorragies et un dépôt d'éosinophiles dans la lumière des
tubules. G2 a montré des tubules presque normaux mais un léger décollement de la
muqueuse épithéliale de quelques tubules et quelques glomérules atrophiés. G3 et
G4 ont des glomérules normaux, des tubules proximaux, distaux et alambiqués.
L'étude de corrélation est réalisée entre les cytokines et les chémokines utilisées et
a montré que G1 a montré une corrélation positive entre IL-10 et IL-6, ainsi
qu'entre CCL2 et VEGF. Cependant, il a montré une corrélation négative entre IL-
10 et CCL2, entre IL-10 et VEGF, entre IL-6 et VEGF, et entre TGF-β1 et CCL2.
G2 ne montre aucune relation de corrélation entre les paramètres utilisés. G3 a
montré une relation de corrélation négative entre TGF β1 et CCL-2, ainsi qu'entre
IL-6 et INF-γ, tandis que le reste des relations entre les critères utilisés n'a pas
montré de relation de corrélation claire. G4 ne montre pas de relation de
corrélation entre les paramètres utilisés, à l'exception de la corrélation entre CCL-5
et INF-γ (corrélation positive), ainsi qu'entre TGF β1 et VEGF (corrélation
négative). Les BM-MSCs ont un rôle thérapeutique essentiel et efficace dans le
traitement du lupus au niveau sérologique en transmettant les cytokines aux
niveaux normaux et au niveau moléculaire en régulant l’expression des gènes et le
niveau histopathologique en traitant les changements histopathologiques
pathologiques pathologiques.
Mots-clés: ELIZA, RT-PCR, souris, cellules souches mésenchymateuses,
cytokines, Balb/c SLE
XVII
Introduction
Introduction
An immune system disorder against the body tissues causes Systemic Lupus
Erythematosus (SLE) or Lupus. Lupus is an inflammatory hereditary chronic
disease. The children inherit their parents' genetic factors predisposing them to
SLE (García-Carrasco et al., 2013).
The causes of Lupus are unknown, but the genetic, environmental, and
hormonal factors greatly affect lupus development. Lupus occurs nine times more
commonly in women than men (Ameer et al., 2022). Lupus appears in all ages and
both genders. The environmental agents include sunlight, medicine, viruses, food,
and chemical substances that have a role in the occurrence of Lupus (Justiz
Vaillant et al., 2023). The main cause of Lupus is not precisely known yet
(Fernandez and Kirou, 2016). Lupus includes inflammation of the skin, joints, and
membranes of the lung, heart, and kidneys, and also development to impair the
immune system (D'Cruz, 2006).
Lupus has six types: SLE, Neonatal Lupus, Chronic Cutaneous Lupus
Erythematosus, Overlap Syndromes, Drug Related Lupus, and Subacute Cutaneous
Lupus (Ameer et al., 2022). Lupus usually includes skin rashes on the face, which
increase after exposure to sunlight (butterfly shape), and rashes can make marks on
other parts of the body (Metry et al., 2019).
Also, lupus symptoms included fatigue, fever, appetite, and weight loss. The
whitening bruising on the fingertips is demonstrated in some patients (Batool et al.,
2016). Lupus affects various organs, such as the brain and spinal cord.
Neurological symptoms can occur in some individuals with Lupus, ranging from
mild to severe. Lupus causes cognitive Dysfunction, Seizures, Psychiatric
Symptoms, Headaches, and Vasculitis. Lupus Patients have pain and swelling of
the joints. The kidney is the important and greater organ that is the target of Lupus,
and nearly 50% of the patients have impaired renal. The edema in the legs is
formed in some patients with protein loss. Some patients may have renal failure
(Justiz et al., 2023).
The classical therapy of lupus disease is to stop the disease progression and
complications. Immunosuppressive and Anti-inflammatory drugs are used against
the inflammations that occur due to Lupus (Kuhn et al., 2015). The long uses and
high doses of Corticosteroids have negative effects on health. The studies show the
side effects of Anti-inflammatory drugs include weight gain due to fluid retention,
1
Introduction
over many years are crucial for understanding the progression of Lupus. Some
biomarkers are associated with Lupus; the search for specific and reliable
biomarkers to aid in diagnosis, prognosis, and treatment response is ongoing. The
medications used to treat Lupus can also introduce confounding variables in
studies. Some studies rely on patient self-reported data, which may introduce recall
bias or be subject to interpretation (Piga et al. 2021).
The main aim of the current work is to treat Lupus by mesenchymal stem
cells in mice, and this aim includes many objectives:
3
Literature
Literature
Politics, religion, ethics, and morality all factor into the widespread fascination
with and discussion of stem cell research. Controversies have arisen regarding
which form of stem cell should be used in the future, given the enormous
adaptability of embryonic stem cells and the unprecedented studies revealing the
plasticity of adult stem cells (Zakrzewski et al., 2019). The human fascination with
creatures that can heal themselves has a long history. The extraordinary
physiological process of regeneration is the organization of neighboring tissues to
replace an injured or severed body section. Despite all the necessary directions for
producing the tissue from the embryonic tissues of the mammals have lost the
regenerative ability, perhaps as a trade-off for more proficient wound healing
ability, compared to several invertebrates like planarian flatworms and Hydra
(Boroviak et al., 2014).
4
Literature
5
Literature
them at increased risk for uncontrolled growth and the development of tumors
(Rippon and Bishop, 2004).
Blastomeric proliferation is necessary for the shift from blastocysts to ESCs,
which affects the blastocysts' cells and genes. The undifferentiated ESCs do not
provoke an immune response upon transplantation; this is not true for more
differentiated cells derived from ESCs, which begin to express major
histocompatibility complex (MHC). MHC matching is indicated when ESCs are
used clinically (Vazin and Freed, 2010), as shown in Figure 1.
6
Literature
7
Literature
WJ-MSCs have many characteristics with embryonic stem cells, both in terms
of their phenotype and genetic makeup (Varaa et al., 2019).
8
Literature
9
Literature
10
Literature
required to produce all blood cell lineages (Mosaad YM, 2014) (Morrison et al.,
1995).
In addition to their capacity for self-renewal, one characteristic that sets HSCs
apart from other mature cells is their capacity to differentiate into cells of various
lineages in a targeted and extensive manner. These results demonstrated HSC
activity and additional measurement of the potential of HSCs to create mature
blood cells in patients after myeloablation was carried out (Hawley et al., 2006).
Similar to LT-HSC, HSC/progenitors may either directly or indirectly contribute to
the process of repairing tissues and treating blood diseases (Khaddour et al., 2023).
HSCs have a high ability to differentiate into mature blood cells. Since the initial
transplantation experiments hinted at their existence over 50 years ago, the
capacity to proactively isolate HSCs has been the topic of intense inquiry (Lee and
Hong, 2020).
Despite significant advances in enrichment protocols, the continuous in vitro
propagation of human HSCs has not yet been achieved. This chapter describes
current procedures used to phenotypically and functionally characterize candidate
human HSCs and initial efforts to derive permanent human HSC lines (Bryder et
al., 2006). Hematopoietic stem cells are considered one of the most important cell
sources when treating degenerative illnesses. The adult hematopoietic stem cells
are used in many therapeutic techniques. It is considered an essential cell source in
stem cell biology. HSCs have been used in clinical and fundamental scientific
research up to this point (Eaves, 2015) (Ng and Alexander, 2017), as shown in
Figure 4.
11
Literature
12
Literature
The single stem cell can undergo asymmetrical cell division, allowing it to
make an identical daughter cell and reproduce. The single stem cell can also
produce a committed progenitor cell, which can later develop into an adult
epithelial cell type. Because of the evidence that they uncovered, they came to this
13
Literature
conclusion (Andersson-Rolf et al., 2017). When treated for fibrosis and Crohn's
disease by mesenchymal diseases, the stem cell has plasticity ability in many
tissues, such as differentiating into intestinal subepithelial myofibroblasts (Liand
jasper, 2016). Many studies are showed that stem cells proliferate into adult cell
lineages of the gut (Quante and wang, 2009). Adult stem cells can survive long and
regenerate large amounts of tissue (Potten, 1998). Although the stem cells of the
gastrointestinal system are among the most diligent in the body owing to the quick
pace of cell turnover in this tissue, they have not been recognized because of their
immature, undifferentiated phenotype because the rate of cell turnover in this
tissue is so high (Brittan and Wright, 2004), as shown in figure 6.
Figure 6: Oral mucosa in mice. A diagram of the buccal mucosa was taken
from the C57BL/6 model. There are several layers (basal, spinous, granular,
and cornified). The epithelial stem cells are below the layers (Jones and Klein,
2013).
14
Literature
Häussinger, 2013). The oval cells may have positive staining for pyruvate kinase
isoenzyme L-PK, albumin, and alpha-fetoprotein in transforming into hepatocytes.
There is mounting evidence that stem cells derived from bone marrow may play a
role in liver regeneration (Alison et al., 2004) (Habeeb et al., 2015). The
hepatocyte transplants in animals have demonstrated that a specific percentage of
hepatocytes are can proliferate, which mean that hepatocytes have same the
functional stem cells. This gives birth to cords of potential so-called oval cells
within the lobules that can develop into hepatocytes and biliary epithelial cells
(Zhang et al., 2003).
16
Literature
Yamanaka named iPSCs with a lowercase "i" due to the popularity of the iPod
and other products. In his Nobel seminar, Yamanaka cited the earlier seminal work
of Harold Weintraub on the role of myoblast determination protein 1 in
reprogramming cell fate to a muscle lineage as an important precursor to the
discovery of iPSCs (Bragança et al., 2019). PPSCs can develop into three primary
cell groups (Abu-Dawud et al., 2018). All developed cell types are precious in
regeneration medicine and organ formation (Worku, 2021) (Zhu and Huangfu,
2013).
17
Literature
Multipotent stem cells can differentiate into all cell types within one particular
lineage. Plentiful advantages and usages exist for multipotent stem cells (Alberts B
et al., 2002). Multipotent stem cells can develop, repair, and protect tissue.
Multipotent Stem cells could be used in treating spinal cord injury, bone fracture,
autoimmune diseases, rheumatoid arthritis, and fertility preservation (Khanlarkhani
et al., 2016). MPSCs are unspecialized cells that can differentiate into body cells
with specific functions. MPSCs in bone marrow can give all blood cells. MSCs can
produce many body cell types (Mirzaei, 2018).
These stem cells can produce only one cell type but have the property of self-
renewal that distinguishes them from non-stem cells. Examples of unipotent stem
cells are germline stem cells and epidermal stem cells. Unipotent stem cells have
the narrowest differentiation capabilities, making them a promising candidate for
therapeutic use in regenerative medicine (Zakrzewski et al., 2019) (Dulak et al.,
2015). Unipotent stem cells are derived from multipotent cells, which can only
become a limited number of cell types. Multipotent stem cells are known to create
bone, muscle, cartilage, fat, and other similar tissues. Unipotent stem cells are
derived from other types of stem cells but, at some point, differentiate them to
become a specific tissue type (De Kretser, 2007).
18
Literature
frequently to treat blood and bone marrow illnesses, blood malignancies, and
immunological problems. Recently, peripheral blood and umbilical cord stem cells
have been employed to treat several blood-based disorders (Duran et al., 2001),
edtiales of the mentioned stem cells in the current study are listed in the following
tables 1.
Table 1: types of the stem cells that mentioned in the current study with
sources
Types of stem cells according to the source
Source The stem cell Abbreviation Collection
name site
Embryos Embryonic ESCs The inner cell
stem cells mass of the
blastocyst
before
implantation
Embryonic EGCs The somatic
Germ Stem lineage
Cells between late
embryonic and
early fetal
development
Fetal stem cells FSC fetal blood and
bone marrow,
fetal liver and
kidney
infant Umbilical cord UCSC Umbilical
stem cells cord
Wharton's jelly WJSC Umbilical
stem cells cord
Adults Mesenchymal MSCs Bone marrow
stem cells and adipose
tissues
Hematopoietic HSC -bone marrow,
stem cells -peripheral
blood,
-umbilical
cord
19
Literature
20
Literature
(Hoang et al., 2022), the genes for the mouse's alpha-globin were replaced with
human alpha-globin genes, and the genes for the beta-globin were changed with
human sickle beta-globin genes (Siham, 2020).
These cells were then implanted into hs/hs mice that had been irradiated. Blood
smears taken from untreated mice revealed an excess of the sickle-shaped cells
characteristic of sickle cell anemia. Blood smears from treated mice revealed a
significantly reduced number of sickle-shaped cells. Additionally, many red blood
cells were seen in the treated mice. It is believed that a similar approach could be
made in human embryonic stem cells to replace genetic defects and test iPS cells
for effective disease-specific medications (Biehl and Russell, 2009). In 2012, a
newspaper in the United Kingdom announced that the patient who had been the
first in the country to get a transplant of embryonic stem cells had regained part of
his central vision. Marcus Hilton is one of the participants in a human clinical
safety study involving two other persons with Stargardt's disease in the United
States. Stargardt's disease is when the retina starts to deteriorate and thin, affecting
the patient's central vision (Watt and Driskell, 2010). Marcus Hilton is one of the
participants in this experiment.
At the time of his diagnosis, Hilton was ten years old and could only see in the
periphery of his field of vision. In January 2012, at age 34, he had a procedure in
which 50,000 retinal pigment epithelium cells were injected into the area behind
the retina in his right eye (Ebrahimi et al., 2021). Tiny plastic fibers seeded with
stem cells taken from the patient's bone marrow have been used to fabricate the
trachea. After being put within a container known as a bioreactor, the tissue-
engineered scaffold was then doused in a solution that made it possible for the cells
to be completely absorbed. According to (Nadig, 2009), the cells continued
proliferating even after the modified trachea was transplanted.
In 2008, a lady whose trachea had collapsed as a result of the problems caused
by TB had the first-ever trachea transplant procedure. In 2013, five years after the
incident, she was in excellent health, had good lung function, and had no
complications related to rejection. A trachea transplant created from the patient's
stem cells was performed on a patient in Eritrea in July 2011 and on a patient in
Baltimore in January 2012 by the same surgeon, Dr. Paolo Macchiarini. The
transplants were performed to replace tracheas that had been damaged by cancer.
These two cancer patients, however, did not make it through their ordeals. The
tracheal transplants have been attempted in a few youngsters (Musial and Gorska-
21
Literature
Ponikowska, 2021) (Fontes and Thomson, 1999). Both human embryonic stem
cells and induced pluripotent stem cells need a significant amount of further
investigation at the fundamental level. It does not seem that these cells are the
same as embryonic stem cells acquired from mice. It seems that embryonic stem
cells from mice proliferate and develop slower than embryonic stem cells from
humans (Mahla RS., 2016).
These cells might be used to treat various conditions, including cancer. Some
early investigations involving the transplantation of fetal neurons into Parkinson's
patients indicate improvement in some, but not all, of the patients, while some
patients have significant side effects. In addition, ethical concerns about the
beginning of life, the beginning of a parent's responsibilities, embryo rights, and
the rights of embryo donors continue to be raised concerning human embryonic
tissue use (Mousaei et al., 2022).
22
Literature
The Lupus Foundation of America estimates that (1.5) million Americans, and
at least five million people worldwide, have a form of Lupus. Lupus strikes mostly
women of childbearing age. However, men, children, and teenagers develop
Lupus, too. Ninety percent (90%) of people living with Lupus are women. Most
people with Lupus develop the disease between the ages of 15 and 44 (Pons-Estel
et al., 2010).
The best estimate based on available data on incidence is 16,000 new cases per
year (Wallace & Hahn, 2013). Systemic lupus erythematosus (SLE) may be more
prevalent among most ethnic groups in the low-and-middle-income countries; still,
23
Literature
26
Literature
Figure 7: clinical signs of cutaneous Lupus in the humans, the lesion placed in
different sites, A: on the back, B: on the lips, C: on the hand, D: on the face
(Internet source).
Dendritic cells process these microparticles and mature and present these as
antigens to T-cells. T-cells, microparticles, and pro-inflammatory cytokines trigger
B-cell activation and autoantibody production. As a result, body tissues lose their
self-tolerance. The most prominent events involving hormonal abnormalities are
due to prolactin and estrogen (Gallo and Gallucci, 2013).
27
Literature
28
Literature
29
Literature
30
Literature
The role of other forms of genetic variation is an exciting new frontier. Some
copy number variants have already been shown to be important for SLE.
Epigenetic variation is clearly involved in the pathogenesis of SLE. Such change
may be the result of environmental exposures and can have a profound impact on
gene expression. These new findings are creating new hypotheses about
mechanisms of disease that may be potential therapeutic targets and will
revolutionize our knowledge of SLE (Ptacek et al., 2008).
In the last few years, we have witnessed tremendous progress in identifying
genetic factors that contribute to the risk of developing SLE. An important
contribution to the progress in the genetics of SLE has been the unprecedented
collaborations among various research groups. Unfortunately, less effort has been
focused on the genetics of LN, but this appears to be changing (Kyogoku et al.,
2004). A recent estimate suggests that most genetic variation identified so far only
explains about 8% of genetic SLE risk. Once association to a region or
polymorphism is established, identifying the true causal variant is complicated and
time-consuming. Establishing the role of a gene in SLE pathogenesis requires
multiple lines of evidence. Clearly, much remains to be done before the etiology of
SLE and LN becomes fully understood. Despite these qualifications, the near-term
potential to understand much of SLE's etiology has never looked so promising
(Tsao and Wu, 2007).
Many of the SLE susceptibility genes have known immune functions and can
be placed into several focal pathways. These pathways highlight the importance of
immune complex clearance, lymphocyte signaling, and the innate immune
response in SLE predisposition. The genes without immunological function may
reveal novel pathways, and other forms of genetic regulation, such as epigenetic
modifications and miRNAs, promise to unveil novel disease mechanisms. The
development of high-throughput methods and parallel analytical strategies are
expected to provide unprecedented novel insights into the genetic factors and
immune pathways that contribute to the pathogenesis of SLE (Pan and Sawalha,
2009).
There are many genes that related with the lupus diseases. These genes
contribute to predisposing factors for the disease occurance. Its are included HLA-
31
Literature
DR2, -DR3, C2, C4, C1q, FCGR2A, FCGR3A, FCGR2B, PDCD1, PTPN22,
IRF5, TYK2, FCGR3B, STAT4, IRAK1, TREX1, MECP2, TNFSF4, CRP,
PTTG1, UBE2L3, PXK, PHRF1, ICA1, NMNAT2, ITGAM, TNFAIP3, BLK,
BANK1, and TNIP1 (Ramos et al., 2010).
4- Urinalysis:
An examination of the urine sample may show an increased protein level or red
blood cells in the urine, which may occur if Lupus has affected the kidneys
(Bessone et al., 2014).
32
Literature
6- Imaging tests:
A- X-ray:
Lupus affects the lungs or heart. A chest X-ray is suggested. An image of the
chest may reveal abnormal shadows that suggest fluid or lung inflammation
(Sudoł-Szopińska et al., 2022).
B- Echocardiogram:
This test uses sound waves to produce real-time images of the heart. It can check
for problems with the valves and other portions of the heart (Mohamed et al.,
2019).
7- Biopsy
Lupus can harm the kidneys in many ways, and treatments can vary depending
on the damage type. In some cases, it's necessary to test a small sample of kidney
tissue to determine what the best treatment might be. The sample can be obtained
with a needle or through a small incision. Skin biopsy is sometimes performed to
confirm a diagnosis of Lupus affecting the skin (Jatwani and Hearth, 2023).
33
Literature
34
Literature
have immunomodulatory effects. Human umbilical cord MSC. may regulate the
aberrant immunological responses of T cells or B cells in SLE. The study found
that hUCMSCs and hUCMSC-EVs increased (Th) 17 cell numbers and IL-17 and
TGF-1 in lupus cases (Xie et al., 2022) (Qiaoli et al., 2023). B6-MSC and lupus-
MSC from young mice suppressed splenic CD3+CD4+ T cells and CD19+CD21+
B lymphocytes in MRL/lpr mice and (NZB/NZW)F1 mice. Lupus-MSC from older
(NZB/NZW) F1 mice did not substantially lower spleen weights, glomerular IgG
deposits, inflammation, CD3+CD4+ T cells, or CD19+CD21+ B lymphocytes (Fei
et al., 2012).
Lupus nephritis causes proteinuria, hypertension, and renal failure in more than
half of patients with SLE. MSC treatment resulted in lower levels of ANA, Scr,
BUN, proteinuria, and renal sclerosis score, and MSC treatment could get higher
albumin levels. The MSC treatment group had lower IL-2, IL-12, IL-17, and IFN-γ
(Zhou et al., 2002). MSCs have immunoregulatory and therapeutic effects on
Lupus. MSCs produce anti-inflammatory cytokines and inhibit the pro-
inflammatory cytokines. IL-1 has immunosuppressive activity that enhances the
therapeutic SLE (Xu et al., 2020). MSCs are considered a promising tool for
treating autoimmune diseases like Lupus. MSCs decreased Th1 and Th17 cells by
stimulating Foxp3 in Treg cells (Park et al., 2015).
Another study found that MSCs taken from SLE patients are defective. It can't
be used for treating Lupus. Normal MSCs increase the survival rate and decrease
the disease activity and autoantibodies level (Wang et al., 2013). MSCs
35
Literature
transplantation inhibiting Akt/GSK3β of T cells in lupus mice (Ji et al., 2012). The
intra-renal injection of MSCs in mice's lupus nephritis showed a decrease in
inflammation (Bukulmez et al., 2021) (Nan et al., 2014). Adipose tissue MSCs
have a high effect in lupus mice at a dose (5×105) IV. MSCs ameliorated improved
Lupus histopathological and serological markers (Choi et al., 2012). MSCs reduced
the glomerular immune complex formation and cell infiltration (Schena et al.,
2010) (Ma et al., 2013).
36
Materials and
Methods
Materials and Methods
Background:
The current work is an experimental study. The study includes four groups, each
comprising ten animals (BalB/c mice).
1- The first group (lupus group) is administered ALD-DNA for lupus
induction.
2- The second group (treated group) is administered ALD-DNA, then treated
by mesenchymal stem cells IV.
3- The third group (positive control) is administered mesenchymal stem cells
(MSCs) in a single dose via IV.
4- The fourth group (negative control) is administrated PBS.
The study aims to treat lupus disease by mesenchymal stem cells in BalB/c
mice.
Table 2: illustrate the used materials and equipment’s in the current work
n The material The origin
1 BALB/c-MSCs CellBiologics company, USA
2 RPMI-1640 Gibco, Ireland
3 fetal bovine serum Gibco, Ireland
4 Mglutamine Sigma, USA
5 penicillin G and streptomycin GIBCO
6 proteinase K Sigma, USA
7 S1 nuclease TaKaRa, Japan
8 DNA extraction kit Genetech, China
9 The nanodrop Thermo Fisher Scientific, USA
10 PBS Sigma, USA
11 chloroform, cotton swap, China
syringe, needles, gel tube
12 ELISA kit antinuclear antibodies (MyBioSource, USA)
(ANA)
13 ELISA kit anti-double strand (MyBioSource, USA)
DNA (Anti-dsDNA)
14 The ELISA kit Interleukin-10 Abcam™ UK
37
Materials and Methods
38
Materials and Methods
39
Materials and Methods
Detailed Description
BALB/c mice are particularly well known for producing plasmacytomas
following injection with mineral oil, forming the basis for the production of
monoclonal antibodies. Although not all BALB/c substrains have been examined
for plasmacytoma induction, substrains derived from the Andervont (An) lineage
(which includes BALB/cByJ) typically are susceptible, while those descended
from BALB/cJ are resistant. BALB/c mice immunized with PLP180-199 develop
an atypical form of experimental autoimmune encephalomyelitis (EAE) in which
location determines susceptibility. BALB/c mice are predisposed to dystrophic
cardiac calcinosis. They use BALB/cByJ and BALB/cJ mice obtained from
Jackson Laboratory. This is a substrain of BALB derived initially from the BALB
strain maintained by MacDowell at Cold Spring Harbor, eventually sent to G.D.
Snell at the University of Texas and brought by Snell to the Jackson Laboratory in
1932 at generation F26.
Cardiovascular Research, Diet-Induced Atherosclerosis, induction autoimmune
disease, lupus, Cancer Research, Mammary Gland Tumors: late onset, Research
Tools, Monoclonal antibodies, Myeloma, Infectious Disease, Neurodevelopmental
Defects, Inflammation, Autoimmunity, Allergic encephalomyelitis, Hematological
Research, Hemoglobin Defects and Thalassemia beta (see Annexes 4).
40
Materials and Methods
1-Preparation of splenocyte:
Removing the BALB/c mice spleens and keeping them in RPMI-1640 (Gibco,
Ireland), then passing the cells several times on nylon mesh, then washing by
RPMI-1640, Add fetal bovine serum (Gibco, Ireland) (10%) with Mglutamine 2m
(Sigma, USA), penicillin G and streptomycin (100) mg/ml each one to prevent the
contamination. Then, splenocytes were diluted to (2×10 6) cells/ml with
concanavalin A (5 ug/ml) for 48 hours.
2-DNA extraction:
gDNA extracted activated splenocytes were treated with proteinase K (Sigma,
USA) and S1 nuclease (TaKaRa, Japan) and then purified by kit (Genetech,
China). The nanodrop (Thermo Fisher Scientific, USA) is used at (260) nm for
determining DNA concentrations.
The immunization:
The mice were classified into four groups. Each consists of ten mice (G1, G2,
G3, and G4). G1 and G2 groups were only immunized with activated ALD DNA
(2000 ug /kg) SC with PBS and (CFA) (Sigma, USA) at three doses of ALD DNA;
the period between one and another was two weeks, at (0, 14, 28) day. Before the
third dose, the animals showed clinical signs of SLE syndrome (nose bleeding), as
shown in Figure 10.
41
Materials and Methods
Figure 10: Steps of preparation of the activated lymphocyte DNA (ALD DNA)
according to (Qiao et al., 2005)
42
Materials and Methods
NC x D x 104/#Q
NC= the cell count
D= the sample dilution
#Q= the squares number of the Neubauer
44
Materials and Methods
paratopes that can be identified by both the pre-coated capture antigen and the
detection antigen simultaneously (see Annexes 2).
45
Materials and Methods
This kit contains materials with small quantities of sodium azide. Sodium azide
reacts with lead and copper plumbing to form explosive metal azides. Upon
disposal, flush drains with a large volume of water to prevent azide accumulation.
Avoid ingestion and contact with eyes, skin, and mucous membranes. In case of
contact, rinse the affected area with plenty of water. Observe all federal, state, and
local regulations for disposal (see Annexes 8).
Species reactivity Reacts with: Mouse Product overview Abcam's Mouse IL-10
ELISA kit is an in vitro enzyme-linked immunosorbent assay for quantitatively
measuring IL-10 in serum. This assay employs an antibody specific for Mouse IL-
10 coated on a 96-well plate. Standards and samples are pipetted into the wells,
and IL-10 present in a sample is bound to the wells by the immobilized antibody.
The wells were washed, and a biotinylated anti-mouse IL-10 antibody was added.
After washing away unbound biotinylated antibodies, HRP-conjugated streptavidin
is pipetted to the wells. The wells are again washed, a TMB substrate solution is
added to the wells, and color develops in proportion to the amount of IL-10 bound.
The Stop Solution changes the color from blue to yellow, and the intensity of the
color is measured at 450 nm (see Annexes 9).
46
Materials and Methods
Sample type Milk, Serum, Assay type Sandwich (quantitative), Assay duration
multiple steps standard assay Species reactivity Reacts with: Mouse Product
overview Mouse interferon gamma ELISA kit (IFN Gamma) is designed for the
quantitative measurement of IFN gamma in mouse serum, buffered solutions, and
cell culture medium. A monoclonal antibody specific for IFN gamma has been
coated onto the wells of the microtiter strips provided. Samples are pipetted into
these wells, including standards of known IFN gamma concentrations, control
specimens, or unknowns. The standards or samples and a biotinylated monoclonal
antibody specific for IFN gamma are simultaneously incubated during the first
incubation. After washing, the enzyme Streptavidin-HRP, which binds the
biotinylated antibody, is added, incubated, and washed. A TMB substrate solution
is added, which acts on the bound enzyme to induce a colored reaction product.
The intensity of this colored product is directly proportional to the concentration of
IFN gamma present in the samples. This kit will recognize both endogenous and
recombinant mouse IFN gamma. Get results in 90 minutes with the Human IFN
gamma ELISA Kit from our Simple Step ELISA range. Platform Microplate
Function is produced by lymphocytes activated by specific antigens or mitogens
(see Annexes 10).
47
Materials and Methods
Quantikine™ ELISA: This package insert must be read in entirety before using
this product. It is not for use in diagnostic procedures but for the quantitative
determination of Mouse VEGF helical Growth Factor concentrations in serum
samples. Vascular endothelial growth factor, or vascular permeability factor,
potentiates both angiogenesis and vasculogenesis in the fetus and adults (see
Annexes 12).
48
Materials and Methods
Collection of organs: The animal's abdomen was opened by scalpel blade and
scissors, and the organs (Liver and Kidney) were excluded and then weighed. After
that, put them in containers to wash them with distal water if it's contaminated with
blood, and then use dissection papers to dry them. After the desiccation of animals,
the organs of the Kidney and Liver are taken away and washed with NS
49
Materials and Methods
physiological solution to remove blood. A small part of Kidney and Liver tissue
for each sample was frozen in liquid nitrogen and kept at −70 °C for molecular
assy. Series steps depending on the method mentioned by Cook (1973) and
Suvarna and Layton (2013) for histological exam include:
1- Fixation: After putting tissues for 48 hours in formalin solution (10%), then
removing and cleaning with continuous water for two hours; this method is called
(Fixation) to save tissue and cells through link protein, prevent shrinking, swelling,
and inactive lysis enzyme.
2- Washing: Washing samples in water for 3 hours to remove the fixative effect on
the samples.
3- Dehydration: Then samples were entered into a graded series of increasing
ethanol concentrations (70%, 80%, 90%, 95%, and 100%) for about 1-2 h. for each
concentration. Using gradual ethanol concentration, the tissue is extracted from
water and transferred from one concentration of alcohol to another.
4- Clearing: The tissue is extracted from alcohol and put in Xylene for 1 hour to get
rid of alcohol to facilitate the next step.
5- Infiltration: Using liquid paraffin at 56-58°C 2 times.
6- Embedding: Then the samples were entered in containers containing liquid
paraffin in 56-58 °C and left in room temperature to be hard, then released from
the containers and put in freeze.
7- Sectioning: the sample is fixed on the sample holder in Rotary Microtome and
cutes to a thick of 5 micron, then the Ribbon is transferred to a water bath (40-45
°C) for float, leaving for (1-2 min). The slide is passed under this Ribbon and
caught so that it sticks to the center of the slide by lifting the slide to word the
sectors to the top while not allowing any air bubbles to form. Leave the slide to dry
on the strip dryer at room temperature for one hour.
8- Deparaffinization and rehydration: Deparaffinization of the slide by putting it in
an oven (55-70 °C) for five h., then in Xylene. Redehydration by putting it in a
graded series of alcohol (100%, 100%, 90%, 80%, 70%) respectively.
9- Staining: Two dyes are used in this study. Hematoxylin, which is prepared,
depends on the method described (Suvarna et al., 2013). The dye powder melted in
absolute alcohol (99%) after adding it to Alk(So4)2-12H2O in warm DW. After
the mix is boiled, red mercury oxide is added to the mixture. The mixture is cooled
by putting it in a beaker with cold water and then adding acetic acid after the
50
Materials and Methods
mixture is filtered for use. Another dye is eosin, which is also prepared depending
on Suvarna and Layton's (2013) method. The dye powder melts in absolute alcohol
(99%) concentration, then adds Glacial Acetic Acid and filters the mixture before
use. The slide was dyed with Hematoxylin for one minute, and after washing the
slide for two minutes, distal water was used to remove the access dye. After that,
twice or three times, it was submerged in alcohol to eliminate access dye. It is
stained with eosin dye and then passed through a series of ethanol concentrations
for (2min) in each concentration (70%, 80%,90%, 100%, 100%) respectively,
except the last concentration for (5 min), after for (10 min) clearing with Xylene.
10- Mounting: Using Distrine Plasticizer Xylene to fix the cover slide, the slide is left
on the plate for (8 hours) to dry and prepare for exams.
1.13. Morphometry
Photomicrograph:
After preparation of sections, slides were examined by Cam Light Microscope,
and the alternative in tissue sections was detected. They were taken on 20X, 40X,
and 100X lenses.
51
Results and
Discussion
Results and Discussion
General background:
The current study found that the lupus group (G1) had a high level of pro-
inflammatory cytokines and increased gene expression. It also decreased the anti-
inflammatory cytokines and decreased its gene expression. The treated group (G2)
has a low level of pro-inflammatory cytokines with a decrease in gene expression
and an increase in anti-inflammatory cytokines with an increase in gene
expression. The histopathological examination of the liver and kidney showed
negative histopathological changes in the liver and kidney in the lupus group,
while the treated group (G2) showed positive histological amelioration of the liver
and kidney tissues.
Our results showed anti-dsDNA levels in the lupus group (G1) were (17.38) ng
/ml. The G1 is left without treatment. Anti-dsDNA levels in the treated group (G2)
before treatment were (17.29) ng/ml, and after treatment was (2.45) ng/ml at
(P≥0.05). The anti-dsDNA levels showed negative significant differences in the
treated group (G2) after treatment as compared with the before treatment at
(P≥0.05). There are no significant differences between Anti-dsDNA levels between
G1 and G2 before treatment at (P<0.05), as shown in Table 3.
52
Results and Discussion
Our results showed that ANA levels in the lupus group (G1) were (18.15) ng
/ml. The G1 is left without treatment. ANA levels in the treated group (G2) before
treatment were (17.55) ng/ml, and after treatment was (2.71) ng/ml at (P≥0.05).
The ANA levels showed negative significant differences in the treated group (G2)
after treatment as compared with the before treatment at (P≥0.05). There are no
significant differences between ANA levels between G1 and G2 before treatment
(P≥0.05), as shown in Table 5.
Table 5: ANA and Anti dsDNA level before and after BM-MSCs therapy in
the lupus group (G1) and treated group (G2)
AB Anti-dsDNA ng/ml
Group Before treatment after treatment
G1 17.38 ±0.83 Aa -
G2 17.29 ±0.77 Aa 2.45 ±0.52 b
LSD 2.9
AB ANA ng/ml
Group Before treatment after treatment
G1 18.15±1.45 Aa -
G2 17.55±0.83 Aa 2.71±0.61 b
LSD 2.1
Different letters mean there is a significant difference at p ≤ 0.05 indicates significantly
different, p ≥ 0.05 indicates no significantly different. The capital letters (A and B) indicate
the vertical comparison and the small letters (a, b) indicate the horizontal comparison.
53
Results and Discussion
The anti-dsDNA and ANA are antibodies the lymphocytes produce against self-
DNA due to poor identification of the exact antigenic components (Giles &
Boackle, 2013) (Bai et al., 2018). Anti-dsDNA and ANA antibodies are increased
in lupus, according to many reports (Rekvig, 2019) (Sohrabian et al., 2019), as
found in our results.
Figure 11: Concentration of IL-10 in study groups, Different letters mean is a significant
difference; the similar letters mean no significant differences. A is mean low in the
significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥ 0.05
indicates no significantly different.
IL-10 has anti-inflammatory effects, is produced at low levels in lupus cases
compared with healthy individuals (Godsell et al., 2016), and is suppressed by
many immunobiological factors secreted in lupus involvement (Yuan et al., 2011).
The group treated by MSC showed an increase in IL-10 cytokine levels due to
MSC effects that are yet unknown (Waterman et al., 2012). Some reports indicated
54
Results and Discussion
that MSC could produce exosomes that directly regulate and control the immune
response (Sharma et al., 2017) (Wang et al., 2020). According to Figure 12, the
first group showed a significant increase (360.89±4.48) ng/ml of IL-6 levels as
compared with the second group (201.14±5.55) ng/ml, the third group
(190.89±3.7c) ng/ml, and the fourth group (191.37±3.26). The first group showed
positive significant differences as compared with the other groups at (P≥0.05)
Figure 12: Concentration of IL-6 in study groups, Different letters mean there is a
significant difference; the similar letters mean no significant differences. A is mean high in
significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥ 0.05
indicates no significantly different.
Increased cytokines are clear in target organs in SLE with their receptors
expressed (Davis et al., 2011). Another study found that cytokines, such as
Interferons (IFN), play the primary role in SLE. Interferons can activate rest
55
Results and Discussion
Figure 13: Concentration of TGF-β1 in study groups, Different letters mean there is a
significant difference; the similar letters mean no significant differences. A is mean low in
the significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥
0.05 indicates no significantly different.
According to Figure 14, the first group showed an increase in significant
differences (162.98±11.48) ng/ml of CCL-2/MCP-1 levels as compared with G2
(52.09±6.15) ng/ml, G3 (48.1±10.2) ng/ml, and G4 (49.4±9.75) ng/ml. There are
no significant differences between the second, third, and fourth groups. The first
group showed positive significant differences compared to the other groups at
(P≥0.05).
56
Results and Discussion
Figure 14: Concentration of CCL-2/MCP-1 in study groups. Different letters mean there is
a significant difference; the similar letters mean no significant differences. A is mean high
in the significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥
0.05 indicates no significantly different.
Figure 15: Concentration of CCL-5 in study groups, Values represent mean ± SD;
Different letters mean there is a significant difference at (P≥0.05); the similar letters mean
no significant differences at (P≥0.05). A is mean high in the significant differences as
compared with B.
57
Results and Discussion
Figure 16: Concentration of VEGF-A in study groups, Values represent mean ± SD;
Different letters mean there is a significant difference at (P≥0.05); the similar letters mean
no significant differences at (P≥0.05). A is mean high in the significant differences as
compared with B.
According to Figure 17, the 1st group showed increased significant
differences (302.9±28.31) ng/ml of ICAM-1 level as compared with the 2nd group
(226.8±11.69) ng/ml, the 3rd group (225.3±6.26) ng/ml, and the 4th group
(225.1±8.33) ng/ml. At the same time, there are no significant differences among
the 2nd, the 3rd, and the 4th group. The first group showed positive significant
differences compared to the other groups at (P≥0.05).
Figure 17: Concentration of ICAM-1 in study groups, Different letters mean there is a
significant difference; the similar letters mean no significant differences. A is mean high in
the significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥
0.05 indicates no significantly different.
58
Results and Discussion
According to Figure 18, the 1st group showed increased significant differences
(64.85±4.10) ng/ml in IFN-γ levels as compared with the 2nd (14.12±2.10) ng/ml,
the 3rd group (30.21±2.33) ng/ml, and the 4th group (30.37±2.85) ng/ml, while the
3rd and the 4th group were showed increased significant differences as compared
with the 2nd group. The first group showed positive significant differences as
compared with the other groups at (P≥0.05)
Figure 18: Concentration of IFN-γ in study groups, Different letters mean is a significant
difference; the similar letters mean no significant differences. A is mean high in the
significant differences as compared with B. p ≤ 0.05 mean significantly different. p ≥ 0.05
indicates no significantly different.
Nowadays, MSC is considered therapy for SLE to improve the clinical signs of
the SLE by inhibiting the activity of Th1, Th17, and B cells and increasing the
activity of Th2 and Treg cells. Treatment by MSC has benefits for enhancing the
immunosuppression of MSCs in SLE. Although MSC therapy is reported
ineffective in some studies, some patients with SLE do not respond to the therapy
due to MSC activity or patient-derived factors (Li et al., 2021).
MSC plays a role in regulating SLE immunity (Yang et al., 2021). MSCs have
immunosuppressive effects on immune activities. Administration of MSCs IV will
ameliorate the clinical signs of SLE. Allogeneic MSCs suppress autoimmunity in
mice and humans (Li et al., 2021). Treatment by MSC for animals with SLE
59
Results and Discussion
The third group does not have significant differences compared with the control
group. We added the third group to confirm that it does not cause a cross-
immunological reaction due to histological and immunological mismatching. This
will support the idea that MSC only has an essential role in SLE and has no role in
healthy animals.
The IL-10 gene expression in G1 was lower than in the control. No significant
differences exist among other groups in IL-10 gene expression, as listed in Table 6
and Figure 19.
60
Results and Discussion
Figure 19: The chart presents the fold change values of the IL-10 gene
The current study showed that the IL-6 encoding gene expression was increased
in G1 than in the control. At the same time, there were no statistical differences
between the other groups, as shown in Table 7 and Figure 20.
61
Results and Discussion
Figure 20: The chart illustrates the fold change values of the IL-6 gene
The findings showed that the TGF-β1 expression gene was reduced in G1 than in
the control. No marked differences among other groups in the TGF-β1 expression
gene exist, as listed in Table 8 and Figure 21.
62
Results and Discussion
Figure 21: The chart presents the fold change values of the TGF-β1 gene
The gene expression of the CCL-2/MCP-1 gene was increased in G1 more than
in the control group. There are no marked differences among other groups in gene
expression of the CCL-2/MCP-1-encoding gene, as listed in Table 9 and Figure 22.
63
Results and Discussion
Figure 22: The chart illustrates the fold change values of the CCL-2/MCP-1
gene
The current study showed that the gene expression of the CCL-5/RANTES-
encoding gene was increased in G1 than in control. There are no marked
differences among other groups in gene expression of the CCL-5/RANTES-
encoding gene, as listed in Table 10 and Figure 23.
64
Results and Discussion
Figure 23: The chart illustrates the fold change values of the CCL-5 gene
The gene expression of the VEGF-A encoding gene was increased in G1 more
than in the control group. There are no marked differences among other groups in
gene expression of the VEGF-A-encoding gene, as shown in Table 11 and Figure
24.
Table 11: Values of the fold change of the VEGF-A encoding gene
65
Results and Discussion
Figure 24: The chart presents the fold change values of the VEGF-A gene
The current study showed that ICAM-1 encoding gene expression was
increased in G1 than in control. There are no marked differences among other
groups in the ICAM-1-encoding gene expression, as shown in Table 12 and Figure
25.
Table 12: Values of the fold change of the ICAM-1 gene in study groups
66
Results and Discussion
Figure 25: The chart presents the fold changes values of ICAM-1 gene
The current study showed that gene expression of the IFN-γ-encoding gene was
increased in G1 than in the control, with no marked differences between the other
groups, as shown in Table 13 and Figure 26.
Table 13: Values of the fold change of IFN-γ-encoding gene in study groups
67
Results and Discussion
Figure 26: The chart illustrates the fold changes values of the IFN-γ gene
TGF-β1 and IL-10 expression was increased in lupus patients compared to the
control group. TGF-β1 and IL-10 are increased in lupus patients. IL-10 and TGF-
β1 inhibit the autoreactive immune cells in lupus patients (Abbasifard et al., 2020).
The protein and mRNA expression levels of IL‑6 and TNF‑α were decreased in
MSCs in Osteoarthritis cases than in the control group using RT-PCR.
Furthermore, IL‑4, IL‑10, and TGF‑β1 levels in MSCs increased in lupus than in
the control group, which showed agreement with our results (Yi et al., 2020).
MSCs produce many bioactive factors, such as cytokines and growth factors,
which provide microenvironment regeneration and inhibit local inflammation.
68
Results and Discussion
MSCs have a potent immunosuppressive effect in vivo (Le et al., 2004). MSCs
decrease IL-10 levels in pulmonary vasculitis in lupus mice. MSCs are strong
immunomodulatory cells investigated in graft versus host disease, lupus, and
ulcerative colitis (Prabowo et al., 2021). MSCs have an immunosuppressive effect
on the immune system (Zanone et al., 2010). The mesenchymal stem cell has
distinct gene expression; MSc has an immunomodulatory role in autoimmune
disease (Jansen et al., 2010) (Tyndall et al., 2007). The genetic exchange between
resident and MSc by the microvesicles showed instrumental in MSc therapeutic
effects (Bruno et al., 2013). The cytokine MSc produces is essential in Lupus
(Stępień-Wyrobiec et al., 2008). MSCs can treat rheumatoid arthritis, lupus, and
type I DM (Rosa et al., 2007). MSc has expressed the surface markers and secreted
factors. Therefore, it is used to treat lupus (Altaner et al., 2013) (Estrela et al.,
2017). All the studies above agreed with our findings and directly supported our
results.
69
Results and Discussion
Figure 27: Liver section with hepatocytes necrosis Figure 28: Liver section with sinusoids dilatation
(black arrow), hydropic degenerated hepatocytes (black arrows) and hepatocytes atrophy (G1).
(blue arrows), and dilated sinusoids (yellow arrows), H&E, 40X.
viable cells (red arrow) (G1). H&E, 100X
Figure 29: Severe hemorrhage, hepatocytes 70 Figure 30: Loss of normal striation of
necrosis, sinusoids dilatation (G1). H&E, 400X. hepatocytes, atrophy and necrosis of hepatocytes
and sinusoids dilatation (G1). H&E, 40X.
Results and Discussion
Figure 31: A- Engorged central vein (CV), loss of normal striation of the hepatocytes (black arrow),
hepatocyte necrosis (blue arrow), and sinusoid dilation (red arrow). H&E, 100X. B- Shows engorged central
vein (CV), loss of normal striation of hepatocytes, severe necrosis of hepatocytes (blue arrow), and dilation of
sinusoids (black arrow) (G1). H&E, 400X.
G2 showed that the liver has mostly normal hepatocytes but also a focal area of
necrosis as well as dilation of sinusoids. The liver shows normal hepatocytes with
prominent nuclei, deeply eosinophilic cytoplasm, and some necrotic hepatocytes,
as shown in Figure 32.
Figure 32: A- Normal liver architecture. H&E, 40X. B- Normal hepatocytes (black arrows), focal area of
necrosis (blue arrow). H&E, 100X. C- Shows regeneration of the nuclei inside the hepatocytes and deeply
eosinophilic cytoplasm; necrotic hepatocytes and sinusoids dilation (G2). H&E, 400X.
71
Results and Discussion
Figure 33: The liver section illustrates normal liver Figure 34: the liver section illustrates normal
architectures. H&E (G3), 40X. liver architectures (G3). H&E, 100X.
The fourth group (control group) showed that the liver has normal liver
architectures and hepatocytes, as shown in Figure 35.
Figure 35: The liver section shows normal hepatocytes. H&E, 400X (control group)
72
Results and Discussion
According to the findings, G1 showed that the liver has severe necrosis of
hepatocytes, hydropic degenerating hepatocytes, and dilated sinusoids. Moreover,
the liver shows severe hemorrhage, loss of normal striation of hepatocytes, atrophy
of hepatocytes, and engorged central vein.
SLE causes organ and tissue damage due to high titer of the autoantibodies and
immune complex, such as the liver. SLE causes liver pathology, such as hepatitis,
jaundice, and severe inflammation of the liver biopsy (Salem et al., 2019). SLE
causes severe liver damage, hepatic failure, elevation of liver enzymes, and
necrosis of hepatocytes (Guo & LÜ, 2018). The hepatic disorders in patients with
lupus are frequent. The immunoglobulin G (IgG) deposes in the liver and damages
liver tissue. The liver injury was associated with SLE in mice. The
histopathological changes in life include liver inflammation, killer cell
accumulation, and macrophages due to hepatic-deposited lupus IgG (Fang et al.,
2018).
Lupus causes liver function disorders, such as liver damage, hepatitis, cirrhosis,
hyperplasic parenchymal and vascular lesions, hepatotoxicity, and dilated sinusoid,
congestion (Bessone et al., 2014). SLE causes pathological changes in many
organs, such as the kidney, intestine, and liver. The histopathological examination
of the lupus liver includes hepatitis, Cholangitis, Cirrhosis, congestion, and liver
cell damage (Shahin et al., 2017). Lupus causes liver enzyme abnormalities and
histopathologic findings, which include hepatitis, fatty liver, portal inflammation,
hemangioma, congestion, and hepatocyte hyperplasia (Grover et al., 2014).
Enlarged liver cells are detected in infants with liver diseases but rarely in the
mature. SLE leads to abnormal liver function, aggregation of the giant cells in the
liver, low liver function, and increased liver enzymes (Cairns and McMahon,
1996). In a study, they included (238) patients with lupus, which found that 33
patients had liver abnormalities such as cirrhosis, chronic hepatitis, granulomatous
hepatitis, and steatosis. At the same time, nine patients demonstrated advanced
stages of lupus. Three patients died from liver failure (Bruce et al., 1980).
A study in Japan carried out on patients with lupus found several
histopathological changes in the liver, such as liver congestion, fatty liver,
inflammation of the blood vessels of the liver, hepatocyte hyperplasia, chronic
persistent hepatitis, cholangitis, nonspecific reactive hepatitis, hemangioma.
Moreover, the development of nodular regenerative hyperplasia occurs in the liver
with lupus (Toshiharu et al., 1992). A study used (47) patients with lupus with liver
73
Results and Discussion
function disorder and tissue abnormalities; hepatitis was most common in the
patients with active lupus, increasing the leucocytes and platelets count (Zheng et
al., 2013).
Based on our findings, the group with lupus treated by MSCs showed that the
liver has mostly normal hepatocytes but also a focal area of necrosis, with slight
dilation of sinusoids. The liver shows normal hepatocytes with prominent nuclei,
deeply eosinophilic cytoplasm, and some necrotic hepatocytes. MSCs used for
treating hepatitis in lupus, MScs, can differentiate into many cell types and have
immunomodulatory effects. MSCs repair liver damage and decrease autoimmune
inflammation. MSCs can promote tissue repair, reducing hepatitis and liver
regeneration (Zhang et al., 2020). MSCs have immunosuppressive properties and
can modulate the immune response by inhibiting the proliferation of immune cells,
such as T and B cells, and reducing the pro-inflammatory cytokine levels. MSCs
suppress the lupus inflammation of the liver. MScs have antifibrotic effects that
reduce liver fibrosis by inhibiting the stellate cells, which are responsible for
excessive collagen deposition (Song et al., 2020). MSCs secrete various factors
and molecules that can promote tissue repair and regeneration. These factors can
stimulate the growth and differentiation of endogenous liver cells, promote
angiogenesis, and modulate the inflammatory response (Han et al., 2022). MSCs
induce proliferation and apoptosis; furthermore, MSCs produce VEGF and CXCL-
12, which have a role in repairing damaged tissue (Dandan et al., 2017). MSCs can
differentiate into many cells, demonstrating a role in immunomodulation and
regenerative therapy. MSCs have a valuable application in lupus treatment. MSCs
can treat lupus, rheumatoid arthritis, multiple sclerosis, inflammatory bowel
disease, and Sjogren's syndrome (Jasim et al., 2022). MSCs have potential in
regenerative medicine, including treating liver damage in lupus. MSCs can
regulate the immune system's response (Shandil et al., 2022). In lupus, the immune
system attacks healthy tissues, including the liver. MSCs can suppress the immune
response, reducing inflammation and preventing liver damage (Huang et al., 2022).
MSCs release many anti-inflammatory molecules that lower the inflammatory
response in the liver. MSCs can secrete the cytokines that stimulate the
regeneration of damaged liver cells (Zuhair, 2022). The above showed data in the
literature that agreed with our results on hepatic histopathological changes due to
lupus; SLE was associated with increased liver diseases, including
histopathological changes, liver function abnormalities, and pathological signs.
74
Results and Discussion
MSCs can differentiate into hepatocyte-like cells and integrate into the existing
liver tissue, enhancing the regenerative process (Wu et al., 2020). In liver damage,
fibrosis may be occurring, affecting liver function. MSCs can reduce fibrosis and
stimulate the extracellular matrix, facilitating tissue repair (Eom et al., 2020).
1.4.2. Kidney:
G1 showed atrophy of glomeruli in the kidney, with severe necrosis of renal
tubules of the kidney, vascular dilatation, obliteration of some renal tubules, and
fibrosis of interstitial tissue between renal tubules; in addition to that, there is
hemorrhage and deposition of eosinophilic materials in the lumen of the renal
tubule, as shown in figure (36-37).
Figure 36: Kidney section with atrophy of glomeruli (black arrow); some glomeruli are normal (yellow
arrows) (G1), H&E, 4X and 400X.
75
Results and Discussion
Figure 37: Severe necrosis of renal tubules of the kidney (blue arrows), vascular dilatation (yellow arrow),
obliteration of some renal tubules (black arrow), fibrosis (pink arrow) of interstitial tissue between renal
tubules, hemorrhage (H), and deposition of eosinophilic materials (green arrow) in the lumen of the renal
tubule (G1) H&E, 400X.
G2 showed almost normal renal tubules of the kidney but slight sloughing of
the epithelial lining of a few renal tubules; in addition, there are normal renal
glomeruli of the kidney and few atrophied glomeruli, as shown in Figure (38-40).
Figure 38: Almost normal renal tubules of the kidney, but sloughing of the epithelial lining of a few renal
tubules (yellow arrows) was also seen (G2), H&E, 400X.
76
Results and Discussion
Figure 39: Normal renal glomeruli of the kidney (black arrows) and few atrophied glomeruli (blue arrow)
were also seen; almost normal renal tubules with slightly sloughed lining epithelium (yellow arrows) of renal
tubules were evident (G2). H&E, 40X and 400X.
Figure 40: Normal renal glomeruli of the kidney (black arrows) and renal tubules, but mild sloughing of
lining epithelium (red arrow) was also evident (G2). H&E, 40X, 100X, and 400X.
77
Results and Discussion
G3, as shown in Figure 41, wherever, G3 showed that kidney tissues have
normal glomeruli and normal proximal, distal, and convoluted tubules, as shown in
Figure 42.
Figure 41: Normal renal glomeruli of the kidney (black arrows) and renal tubules in (G3). H&E, 400X.
The fourth group showed that kidney tissues have normal glomeruli and normal
proximal, distal, and convoluted tubules, as shown in Figure 42.
Figure 42: Normal renal glomeruli and renal tubules of the kidney (G4) (control group), H&E, 100X and
400X
78
Results and Discussion
According to our results, the lupus group showed atrophy of glomeruli in the
kidney, severe necrosis of renal tubules of the kidney, vascular dilatation,
obliteration of some renal tubules, fibrosis of interstitial tissue of the renal tubules,
in addition to that, there are hemorrhage and deposition of eosinophilic materials in
the lumen of the renal tubule. The lupus group and treated by the MSCs group
showed normal renal tubules of the kidney but slight sloughing of the epithelial
lining of a few renal tubules; in addition, there are normal renal glomeruli of the
kidney and few atrophied glomeruli.
Lupus renal has developed renal failure and renal damage over time. Lupus
nephritis occurs as glomerulonephritis, acute tubule-interstitial damage. Lupus
nephritis showed different degrees of renal failure, depending on the disease
period, disease degree, and glomerular lesions. The advanced stage of lupus
nephritis develops into chronic renal failure and renal damage (Davidson, 2016).
More than three hundred renal biopsies in patients with lupus showed diffuse
proliferative glomerulonephritis, severe renal lesions, nephritis, glomerular lesions,
tubular atrophy, sclerosis, and interstitial fibrosis (Dimitrijević et al. 2002).
The kidney with lupus showed that lesions formed and deposited the immune
complex in the renal glomeruli, tubulo-interstitium, and vessels (Isaac, 2021).
Lupus kidney demonstrated renal fibrinous necrosis, fibrous crescents, and
79
Results and Discussion
The reviewed studies support and agree with our findings that the mesenchymal
stem cells can repair and restore the histopathological changes to the normal
architecture due to their ability to repair damaged tissues due to inflammation
caused by lupus erythematosus.
According to our results, there is a positive correlation between IL-10 and IL-6
at a significant level (0.01) and between CCL2 and VEGF at a significant level (P≥
0.05), wherever R-value was (0.903) and (0.666) respectively in the first group.
However, there is a negative correlation between IL-6 and VEGF, also between
TGF- β1 and CCL2, and finally between CCL-5 RANTES and ICAM-1 at (P≥
0.05), wherever R values were (-0.738), (-0.691), and (-0.653) respectively in the
first group, as shown in Table 14.
Table 14: The correlation relationship between the cytokines and chemokines
used in the first group (G1) by using liner R Pearson tests
CCL-5
IL-10 IL-6 TGF- β1 CCL2 RANTES VEGF ICAM-1 IFN- γ
IL-10 1
IL-6 0.903** 1
TGF-B 0.301 0.325 1
CCL2 -0.503 -0.433 -0.691* 1
CCL-5 -0.425 -0.374 -0.102 -0.209 1
VEGF -0.569 -0.738* -0.445 0.666* 0.109 1
ICAM-1 0.126 0.022 0.267 0.084 -0.653* 0.043 1
IFN-Y 0.005 0.146 -0.014 0.301 0.055 0.191 -0.463 1
* Correlation is significant at the 0.05 level, ** Correlation is significant at the 0.01 level
81
Results and Discussion
antibodies and IL-6, IL-10, and TNF-α levels (Jin et al., 2021). A study designed to
establish the correlation between IL-6 and the severity of SLE found a direct
correlation between the level of IL-6 and the severity of SLE (Ali, 2023).
Another study on cytokine levels in SLE patients found that IL-6, IL-10, IL-12,
and IFN-gamma were significantly elevated in active SLE patients compared to
healthy controls (Chun et al., 2007). A study found that TGF-β1 levels were lower
in SLE patients with SLEDAI scores 1-10 and SDI > 3 and were correlated with
chemokine CCL2 (Becker-Merok et al., 2020). The chemokine CCL5, also known
as RANTES (Regulated upon Activation, Normal T cell Expressed, and Secreted),
has been studied in the context of systemic lupus erythematosus (SLE). Elevated
serum levels of circulating RANTES have been described in SLE patients, and it
has been found that the expression of RANTES mRNA in the urinary sediment is
elevated in patients with active lupus nephritis (LN). One study reported that
RANTES correlates with proteinuria and SLE Disease Activity Index (SLEDAI)
levels, but it is not a good predictor of renal flare and has no correlation with the
activity index, chronicity index, or any histopathological characteristics (Aragón et
al., 2020).
On the other hand, ICAM-1 is a cell surface glycoprotein ligand for the
leukocyte integrin LFA-1. Treatment with cyclosporine has been shown to inhibit
ICAM-1 expression. While information is available on the individual roles of
RANTES and ICAM-1 in SLE, the specific correlation between RANTES and
ICAM-1 in the context of lupus is not well-documented in the provided search
results. Further research may be needed to explore the potential correlation
between these two factors in SLE (Jacob et al., 2011).
82
Results and Discussion
The results of the current study showed no correlation relationship between any
of the parameters used in the second group, as shown in Table 15.
Table 15: The correlation relationship between the cytokines and chemokines
used in the first group (G2) by using liner R Pearson tests
VEGF-
IL-10 IL-6 TGF- β1 CCL-2 CCL-5 A ICAM-1 INF - γ
IL-10 1
IL-6 -0.419 1
TGF β1 -0.179 0.112 1
CCL-2 0.145 -0.479 0.372 1
CCL-5 -0.128 -0.151 -0.125 0.336 1
VEGF-
A 0.231 0.245 -0.132 -0.003 -0.333 1
ICAM-1 -0.441 0.502 0.008 -0.529 0.335 -0.437 1
INF - γ 0.433 -0.663 0.302 0.344 -0.012 -0.338 -0.225 1
Based on the results, the results demonstrated that negative correlation between
IL-6 and INF- γ, also between TGF β1 and CCL-2 at a significant level (P≥0.05),
wherever R values were (-0.736) and (-0.782), respectively in the third group, as
shown in Table 16.
Table 16: The correlation relationship between the cytokines and chemokines
used in the first group (G3) by using liner R Pearson tests
IL-10 IL-6 TGF β1 CCL-2 CCL-5 VEGF ICAM-1 INF- γ
IL-10 1
IL-6 -0.024 1
TGF β1 0.378 -0.036 1
CCL-2 -0.442 -0.136 -0.782* 1
CCL-5 0.137 -0.262 -0.578 0.321 1
VEGF -0.522 -0.105 -0.008 0.138 -0.432 1
ICAM-1 0.110 -0.406 0.517 -0.267 -0.275 0.024 1
INF- γ 0.039 -0.736* -0.203 0.298 0.359 0.081 0.584
* Correlation is significant at the 0.05 level
According to the search results, the interplay between IFN-γ and IL-6 regulates
inflammation and gene expression. IFN-γ controls PMN infiltration and modulates
IL-6 signaling through its soluble receptor to promote their apoptosis and clearance
(McLoughlin et al., 2003). Both IFN-γ and IL-6 are required for increased
83
Results and Discussion
IRF-1 is also involved in the IFN-γ regulated expression of IL-6 with TNF-α
activated p65-NF-κB or via co-activation of TLR3 and TLR7 pathways. IFN-γ
may also drive IL-6 production in combination with IL-1β. Previous studies have
indicated that IFN-γ and IL-6 have opposing roles in cell proliferation, apoptotic
death, and inflammation, regulated by a complex crosstalk mechanism (Zhang et
al., 2022) (Qi et al., 2013). The search results did not provide direct information
about the correlation between TGF-β1 and CCL-2. However, they discussed the
role of the CLC-2 chloride channel in TGF-β1-induced processes and signaling
pathways (Sun et al., 2020) (Wang & Wang, 2013). The results indicated that
TGF-β1 can influence the expression of CLC-2 and CLC-3 chloride channels and
their association with various cellular processes and signaling pathways. While the
search results did not directly address the correlation between TGF-β1 and CCL-2,
they highlighted the involvement of TGF-β1 in cellular processes that may be
related to CCL-2 expression and function. Further research on the relationship
between TGF-β1 and CCL-2 would be needed to determine their direct correlation
(Cheng et al., 2007) (Sun et al., 2016).
The current results reveal a negative correlation between TGF β1 and VEGF,
also between CCL-5 and INF- γ at significant levels (P≥0.05), wherever R values
were (-0.513) and (0.530) respectively in the fourth group, as shown in Table 17.
Table 17: The correlation relationship between the cytokines and chemokines
used in the first group (G4) by using liner R Pearson tests
IL-10 IL-6 TGF β1 CCL-2 CCL-5 VEGF ICAM-1 INF - γ
IL-10 1
IL-6 0.102 1
TGF β1 -0.213 0.398 1
CCL-2 0.489 -0.009 0.042 1
CCL-5 -0.297 -0.335 -0.347 -0.379 1
VEGF -0.275 -0.213 -0.513* -0.493 0.067 1
ICAM-1 -0.502 0.110 0.278 -0.491 0.088 0.045 1
INF- γ -0.392 -0.269 0.095 -0.126 0.530* -0.182 0.223 1
* Correlation is significant at the 0.05 level
84
Results and Discussion
TGF-β1 is known to induce angiogenesis in vitro and in vivo, and it has been
proposed that TGF-β1 induces angiogenesis through an indirect mechanism by
inducing the expression of VEGF and/or other angiogenic factors in epithelial or
other cell types. TGF-β1 induces vascular endothelial cell expression of VEGF,
which mediates the apoptotic activity of TGF-β1 through activation of VEGFR2.
VEGF and TGF-β1 are often co-expressed in tissues where angiogenesis occurs,
notably in various tumors. However, the interactions between these two growth
factors at the level of endothelial cells are not yet fully understood (Ferrari et al.,
2009). In a study on patients with advanced non-small cell lung cancer, serum
VEGF-A, TGF-1β, and IL-1β levels were found to be associated with clinical
outcomes (Kim et al., 2013).
In the end, Through the above study and the results reached and discussed in this
chapter, it may be clear to everyone how important this study is in supporting
research projects in treating and diagnosing lupus. Eight important cytokines were
studied, and previous research indicated their importance and close relationship
with lupus disease. For the first time, the determination of the gene expression of
the genes encoding the studied cytokines was addressed. The results showed gene
expression consistent with and supportive of what was stated in the first serological
part. We confirm that studying the relationship of gene expression to cytokines in
Lupus disease was reported for the first time.
The third and final part of the study included a study of the histopathological
changes in kidney and liver tissue. The results supported what was stated in the
85
Results and Discussion
first two chapters, the first chapter (serological) and the second chapter
(molecular). The third chapter represents the final part of the story concerning the
experimental aspect, and the results came in a dramatic way that supports and
reinforces the results of the first and second chapters.
We hope this study will be part of the scientific studies of the scientific
community interested in studying lupus disease as a diagnosis and treatment.
86
Conclusion and
Perspectives
Conclusion and Perspective
87
Conclusion and Perspective
88
Conclusion and Perspective
89
Conclusion and Perspective
12- Supporting research teams and researchers in all universities around the
world morally, materially, and scientifically with the methods and
techniques used in developing the science related to stem cells in terms of
development, investment, applications, and marketing.
90
References
The references
The references:
91
The references
93
The references
94
The references
97
The references
Fang, Xiang, Muhammad Haidar Zaman, Xuanxuan Guo, et al. (2018). Role
of hepatic deposited immunoglobulin G in the pathogenesis of liver
damage in systemic lupus erythematosus. Fron. in Imm.9: 1457.
Fathollahi A, Gabalou NB, Aslani S. (2018). Mesenchymal stem cell
transplantation in systemic lupus erythematous, a mesenchymal stem
cell disorder. Lupus.;27(7):1053-1064.
https://doi:10.1177/0961203318768889
Fatoye, F., Gebrye, T., & Mbada, C. (2022). Global and regional prevalence
and incidence of systemic lupus erythematosus in low-and-middle
income countries: a systematic review and meta-analysis. Rheumatology
International, 42(12), 2097-2107.
Fava A, Petri M. (2019). Systemic lupus erythematosus: Diagnosis and clinical
management. J. Autoimmun.;96:1-13.
https://doi:10.1016/j.jaut.2018.11.001.
Fayyaz A, Igoe A, Kurien BT, Danda D, James JA, Stafford HA, Scofield RH.
(2015). Haematological manifestations of lupus. Lupus Sci Med. 3;2(1):
https://doi:10.1136/lupus-2014-000078.
Fei Gu, Ivan Molano, Philip Ruiz, Lingyun Sun, Gary S. Gilkeson, (2012).
Differential effect of allogeneic versus syngeneic mesenchymal stem cell
transplantation in MRL/lpr and (NZB/NZW)F1 mice, Clinical
Immunology, Volume 145, Issue 2, P:142-152,
https://doi.org/10.1016/j.clim.2012.08.012.
Fernandez D, Kirou KA. (2016). What Causes Lupus Flares? Curr Rheumatol
Rep.;18(3):14. https://doi:10.1007/s11926-016-0562-3.
Ferrari G, Cook BD, Terushkin V, Pintucci G, Mignatti P. (2009).
Transforming growth factor-beta 1 (TGF-beta1) induces angiogenesis
through vascular endothelial growth factor (VEGF)-mediated
apoptosis. J Cell Physiol. ;219(2):449-58. https://doi:10.1002/jcp.21706.
Ferucci ED, Johnston JM, Gaddy JR, Sumner L, Posever JO, Choromanski
TL, Helmick CG (2014) Prevalence and incidence of systemic lupus
erythematosus in a population-based registry of American Indian and
Alaska Native people, 2007–2009. Arthritis Rheumatol 66(9):2494–2502
Fessel WJ (1974) Systemic lupus erythematosus in the community: incidence,
prevalence, outcome, and first symptoms; the high prevalence in black
women. Arch Intern Med 134(6):1027–1035
Flor M, Weiß M, Selhorst T, et al. (2020). Comparison of Bayesian and
frequentist methods for prevalence estimation under misclassification.
BMC Public Health;20:1135.
98
The references
Fontes PA, Thomson AW. (1999). Stem cell technology. Interview by Abi
Berger. BMJ. 13;319(7220):1308.
https://doi:10.1136/bmj.319.7220.1308.
Forbes S, Vig P, Poulsom R, et al. (2012). Hepatic stem cells. J Pathol.
;197(4):510-8. Erratum in: J Pathol.;226(3):e1.
https://doi:10.1002/path.1163.
Forraz N, McGuckin CP. (2011). The umbilical cord: a rich and ethical stem
cell source to advance regenerative medicine. Cell Prolif. ;44 Suppl
1(Suppl 1):60-9. https://doi:10.1111/j.1365-2184.2010.00729.x.
Gallo PM, Gallucci S. (2013). The dendritic cell response to classic, emerging,
and homeostatic danger signals. Implications for autoimmunity. Front
Immunol. 10;4:138. https://doi:10.3389/fimmu.2013.00138.
García-Carrasco M, Mendoza Pinto C, Solís Poblano JC,. (2013). Systemic
lupus erythematosus. In: Anaya JM, Shoenfeld Y, Rojas-Villarraga A,
et al., editors. Autoimmunity: From Bench to Bedside [Internet]. Bogota
(Colombia): El Rosario University Press; 18. Chapter 25. Available
from: https://www.ncbi.nlm.nih.gov/books/NBK459474/
Geneva-Popova MG, Popova-Belova SD, Gardzheva PN, Kraev KI. A Study
of IFN-α-Induced Chemokines CCL2, CXCL10 and CCL19 in Patients
with Systemic Lupus Erythematosu. Life (Basel). 2022 Feb 8;12(2):251.
doi: 10.3390/life12020251.
Ghazimoradi MH, Khalafizadeh A, Babashah S. (2022). A critical review on
induced totipotent stem cells: Types and methods. Stem Cell Res.
;63:102857. https://doi:10.1016/j.scr.2022.102857.
Giannakakis, K., Faraggiana, T. (2011). Histopathology of Lupus Nephritis.
Clinic Rev Allerg Immunol 40, 170–180. https://doi.org/10.1007/s12016-
010-8207-1
Godsell, J, Rudloff, I, Kandane-Rathnayake, R, et al. (2016).Clinical
associations of IL-10 and IL-37 in systemic lupus erythematosus. Sci.
Rep., 6, 34604. https://doi.org/10.1038/srep34604
Golchin A, Chatziparasidou A, Ranjbarvan P, Niknam Z, Ardeshirylajimi A.
(2021). Embryonic Stem Cells in Clinical Trials: Current Overview of
Developments and Challenges. Adv Exp Med Biol.;1312:19-37.
https://doi:10.1007/5584_2020_592.
Gómez-Bernal F, Quevedo-Abeledo JC, García-González M, Fernández-
Cladera Y, González-Rivero AF, de Vera-González A, Martín-González
C, González-Gay MÁ, Ferraz-Amaro I. Serum Levels of Transforming
Growth Factor Beta 1 in Systemic Lupus Erythematosus Patients.
Biomolecules. 2022 Dec 29;13(1):73. doi: 10.3390/biom13010073.
99
The references
Hongyan, L., Yi, Z., Bao, D., Yuewu, L., & Juan, M. (2013). A study on
clinical and pathologic features in lupus nephritis with mainly IgA
deposits and a literature review. Clinical and Developmental
Immunology.
Huang, Y.; Wu, Q.; Tam, P.K.H. (2022). Immunomodulatory Mechanisms of
Mesenchymal Stem Cells and Their Potential Clinical Applications. Int.
J. Mol. Sci. 23, 10023. https://doi.org/10.3390/ijms231710023
Hulya Bukulmez and Gurinder Kumar (2021). Clinical Use of Mesenchymal
Stem Cells in Treatment of Systemic Lupus Erythematosus, book,
IntechOpen, https://doi:10.5772/intechopen.97261
Idborg, H, & Oke, V. (2021). Cytokines as Biomarkers in Systemic Lupus
Erythematosus: Value for Diagnosis and Drug Therapy. Int. J. Mol.
Sci.; 22(21), 11327. https://doi.org/10.3390/ijms222111327
Imran S, Thabah MM, Azharudeen M, et al. (2021). Liver Abnormalities in
Systemic Lupus Erythematosus: A Prospective Observational Study.
Cureus. 16;13(6):e15691. https://doi:10.7759/cureus.15691.
Isaac Ely Stillman, Chapter 43 - The pathology of lupus nephritis, Editor(s):
George C. Tsokos, Systemic Lupus Erythematosus (Second Edition),
Academic Press, 2021, P:389-413, https://doi.org/10.1016/B978-0-12-
814551-7.00043-X.
Ishii T, Eto K. (2014). Fetal stem cell transplantation: Past, present, and
future. World J Stem Cells. 26;6(4):404-20.
https://doi:10.4252/wjsc.v6.i4.404.
Jacob, N., Stohl, W. (2011). Cytokine disturbances in systemic lupus
erythematosus. Arthritis Res Ther 13, 228.
https://doi.org/10.1186/ar3349
Jansen, B. J., Gilissen, C., Roelofs, H., et al. (2010). Functional differences
between mesenchymal stem cell populations are reflected by their
transcriptome. Stem cells and development, 19(4), 481-490.
Jasim, S.A., Yumashev, A.V., Abdelbasset, W.K. et al. (2022). Shining the
light on clinical application of mesenchymal stem cell therapy in
autoimmune diseases. Stem Cell Res Ther 13, 101.
https://doi.org/10.1186/s13287-022-02782-7
Jatwani S, Hearth Holmes MP. (2023). Subacute Cutaneous Lupus
Erythematosus. [Updated 2023 Jul 31]. In: StatPearls [Internet].
Treasure Island (FL): StatPearls Publishing;.
https://www.ncbi.nlm.nih.gov/books/NBK554554/
Ji S, Guo Q, Han Y, Tan G, Luo Y, Zeng F: (2012).Mesenchymal Stem Cell
Transplantation Inhibits Abnormal Activation of Akt/GSK3β Signaling
101
The references
102
The references
103
The references
Lee, Hong Kyung, Hyung Sook Kim, et al. (2017). CCL2 deficient
mesenchymal stem cells fail to establish long-lasting contact with T cells
and no longer ameliorate lupus symptoms. Scien. Reports 7, no. 1: 1-12.
Lenkiewicz AM. (2019). Epidermal Stem Cells. Adv Exp Med Biol.;1201:239-
259. https://doi:10.1007/978-3-030-31206-0_12.
Leong P-Y, Huang J-Y, Chiou J-Y, et al. (2021). The prevalence and incidence
of systemic lupus erythematosus in Taiwan: a nationwide population-
based study. Sci Rep;11:5631.
Lerang K, Gilboe I, Garen T, Thelle DS, Gran JT (2012). High incidence and
prevalence of systemic lupus erythematosus in Norway. Lupus
21(12):1362–1369
Lewandowski LB, Schanberg LE, Thielman N, Phuti A, Kalla AA, Okpechi I,
Scott C (2017). Severe disease presentation and poor outcomes among
pediatric systemic lupus erythematosus patients in South Africa. Lupus
26(2):186–194
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. (2021).Mesenchymal
Stem Cell Therapy: Hope for Patients With Systemic Lupus
Erythematosus. Front Immunol. 30;12:728190.
https://doi:10.3389/fimmu.2021.728190.
Li H, Jasper H. (2016). Gastrointestinal stem cells in health and disease: from
flies to humans. Dis Model Mech. 1;9(5):487-99.
https://doi:10.1242/dmm.024232.
Li Z, Hu X, Zhong JF. (2019). Mesenchymal Stem Cells: Characteristics,
Function, and Application. Stem Cells Int.,6;2019:8106818.
https://doi:10.1155/2019/8106818.
Li, W, Chen, W, Sun, L. (2021). An update for mesenchymal stem cell therapy
in lupus nephritis. Kidney Dis.,;7(2), 79-89.
https://doi:10.1159/000513741
Liang, Jun, Huayong Zhang, Bingzhu Hua, et al. (2010). Allogenic
mesenchymal stem cells transplantation in refractory systemic lupus
erythematosus: a pilot clinical study. Annals of the rheumatic diseases
69, no. 8: 1423-1429.
Liao, Y. W., Chen, Y. M., Hsieh, T. Y., Hung, W. T., Hsu, C. Y., Wen, M. C.,
... & Huang, W. N. (2023). Renal Histopathology Associated With
Kidney Failure and Mortality in Patients With Lupus Nephritis: A
Long-Term Real-World Data Study. The Journal of rheumatology,
jrheum-220345.
Liau LL, Ruszymah BHI, Ng MH, Law JX. (2020). Characteristics and
clinical applications of Wharton's jelly-derived mesenchymal stromal
104
The references
105
The references
Mao, F., Xu, W. R., Qian, H., Zhu, W., et al. (2010). Immunosuppressive
effects of mesenchymal stem cells in collagen induced mouse arthritis.
Inflammation Research, 59, 219-225.
Marino L, Castaldi MA, Rosamilio R, et al. (2019). Mesenchymal Stem Cells
from the Wharton's Jelly of the Human Umbilical Cord: Biological
Properties and Therapeutic Potential. Int J Stem Cells. 31;12(2):218-
226. https://doi:10.15283/ijsc18034.
McKeon KP, Jiang SH. (2020). Treatment of systemic lupus erythematosus.
Aust Prescr. ;43(3):85-90. https://doi:10.18773/austprescr.2020.022.
McLoughlin RM, Witowski J, Robson RL,et al. (2003). Interplay between
IFN-gamma and IL-6 signaling governs neutrophil trafficking and
apoptosis during acute inflammation. J Clin Invest.;112(4):598-607.
https://doi:10.1172/JCI17129
Memon B, Abdelalim EM. (2020). Stem Cell Therapy for Diabetes: Beta Cells
versus Pancreatic Progenitors. Cells., 23;9(2):283.
https://doi:10.3390/cells9020283.
Metry AM, Al Salmi I, Al Balushi F, et al. (2019). Systemic Lupus
Erythematosus: Symptoms and Signs at Initial Presentations.
Antiinflamm Antiallergy Agents Med Chem.;18(2):142-150.
https://doi:10.2174/1871523018666181128161828.
Mingxia Shi, Jing Li, Lianming Liao, Bin Chen, et l. (2007). Regulation of
CXCR4 expression in human mesenchymal stem cells by cytokine
treatment: role in homing efficiency in NOD/SCID mice.
Haematologica; 92(7):897-904;
Mirzaei H, Sahebkar A, Sichani LS, et al. (2018). Therapeutic application of
multipotent stem cells. J Cell Physiol.;233(4):2815-2823.
https://doi:10.1002/jcp.25990.
Mitalipov S, Wolf D. (2009). Totipotency, pluripotency and nuclear
reprogramming. Adv Biochem Eng Biotechnol.;114:185-99.
https://doi:10.1007/10_2008_45.
Mohamed AAA, Hammam N, El Zohri MH, Gheita TA. (2019). Cardiac
Manifestations in Systemic Lupus Erythematosus: Clinical Correlates
of Subclinical Echocardiographic Features. Biomed Res Int.
10;2019:2437105. https://doi:10.1155/2019/2437105.
Mok CC, Lau CS. (2003). Pathogenesis of systemic lupus erythematosus. J
Clin Pathol.;56(7):481-90. https://doi:10.1136/jcp.56.7.481.
Morasso MI, Tomic-Canic M. (2005). Epidermal stem cells: the cradle of
epidermal determination, differentiation and wound healing. Biol
Cell.;97(3):173-83. https://doi:10.1042/BC20040098
106
The references
Morgani SM, Canham MA, Nichols J, Sharov AA, Migueles RP, Ko MS,
Brickman JM. (2013). Totipotent embryonic stem cells arise in ground-
state culture conditions. Cell Rep. 27;3(6):1945-57.
https://doi:10.1016/j.celrep.2013.04.034.
Morrison SJ, Uchida N, Weissman IL. (1995). The biology of hematopoietic
stem cells. Annu Rev Cell Dev Biol.;11:35-71.
https://doi:10.1146/annurev.cb.11.110195.000343.
Mosaad YM. (2014). Hematopoietic stem cells: an overview. Transfus Apher
Sci.;51(3):68-82. https://doi:10.1016/j.transci.2014.10.016.
Mousaei Ghasroldasht M, Seok J, Park HS, et al. (2022). Stem Cell Therapy:
From Idea to Clinical Practice. Int J Mol Sci. 5;23(5):2850.
https://doi:10.3390/ijms23052850
Murtaugh LC, Kopinke D. Pancreatic stem cells. (2008). In: StemBook
[Internet]. Cambridge (MA): Harvard Stem Cell Institute;
https://doi:10.3824/stembook.1.3.1
Musial C, Gorska-Ponikowska M. (2012). Medical progress: Stem cells as a
new therapeutic strategy for COVID-19. Stem Cell Res. ;52:102239.
https://doi:10.1016/j.scr.2021.102239.
Nadig RR. (2009). Stem cell therapy - Hype or hope? A review. J Conserv
Dent.;12(4):131-8. https://doi:10.4103/0972-0707.58329
Nan Che, Xia Li, Lu Zhang, Rui Liu, Haifeng Chen, Xiang Gao, Songtao Shi,
Wanjun Chen, Lingyun Sun; (2014). Impaired B Cell Inhibition by
Lupus Bone Marrow Mesenchymal Stem Cells Is Caused by Reduced
CCL2 Expression. J Immunol; 193 (10): 5306–5314.
https://doi.org/10.4049/jimmunol.1400036
National Research Council (US) and Institute of Medicine (US) (2002).
Committee on the Biological and Biomedical Applications of Stem Cell
Research. Stem Cells and the Future of Regenerative Medicine.
Washington (DC): National Academies Press (US); CHAPTER THREE,
Embryonic Stem Cells.
https://www.ncbi.nlm.nih.gov/books/NBK223690/
Ng AP, Alexander WS. (2017). Haematopoietic stem cells: past, present and
future. Cell Death Discov. 6;3:17002.
https://doi:10.1038/cddiscovery.2017.2.
Nyman E, Vaughan T, Desta B, Wang X, Barut V, Emmas C. (2020).
Characteristics and Symptom Severity of Patients Reporting Systemic
Lupus Erythematosus in the PatientsLikeMe Online Health
Community: A Retrospective Observational Study. Rheumatol
Ther.;7(1):201-213. https://doi:10.1007/s40744-020-00195-7.
107
The references
Poole, BD, Niewold, TB, Tsokos, GC, & Via, CS. (2010). Cytokines in systemic
lupus erythematosus. J. biotechnol. biomed., 735169.
https://doi.org/10.1155/2010/735169
Potten CS. (1998). Stem cells in gastrointestinal epithelium: numbers,
characteristics and death. Philos Trans R Soc Lond B Biol Sci.
29;353(1370):821-30. https://doi:10.1098/rstb.1998.0246
Prabowo, N. A., Adnan, Z. A., et al. (2021). Mesenchymal stem cell
conditioned medium as good as methyl prednisolone in decreasing levels
of interleukin 10 and the degree of pulmonary vasculitis in lupus mice.
Bangladesh Journal of Medical Science, 20(2), 426-430.
Ptacek T, Li X, Kelley JM, Edberg JC. (2008). Copy number variants in
genetic susceptibility and severity of systemic lupus erythematosus.
Cytogenet Genome Res. ;123:142–147.
Qi, Yf., Huang, Yx., Wang, Hy. et al. (2013). Elucidating the crosstalk
mechanism between IFN-gamma and IL-6 via mathematical modelling.
BMC Bioinformatics 14, 41. https://doi.org/10.1186/1471-2105-14-41
Qiao B, Wu J, Chu YW, Wang Y, et al. (2005). Induction of systemic lupus
erythematosus-like syndrome in syngeneic mice by immunization with
activated lymphocyte-derived DNA. Rheumatology. Sup; 44(9):1108-14.
https://doi:10.1093/rheumatology/keh656
Qiaoli Yi, Zhijie Xu, Abhimanyu Thakur, Kui Zhang, Qiuju Liang, Yuanhong
Liu, Yuanliang Yan. (2023). Current understanding of plant-derived
exosome-like nanoparticles in regulating the inflammatory response and
immune system microenvironment. Pharmacological Research 190,
pages 106733.
Qinjun Zhao, Hongying Ren, Zhongchao Han, (2016). Mesenchymal stem
cells: Immunomodulatory capability and clinical potential in immune
diseases, Journal of Cellular Immunotherapy, V:2, Issue 1, P:3-20,
https://doi.org/10.1016/j.jocit.2014.12.001
Quante M, Wang TC. (2009). Stem cells in gastroenterology and hepatology.
Nat Rev Gastroenterol Hepatol.;6(12):724-37.
https://doi:10.1038/nrgastro.2009.195.
Quintero-Del-Rio AI, Kelly JA, Kilpatrick J, et al. (2002). The genetics of
systemic lupus erythematosus stratified by renal disease: linkage at
10q22.3 (SLEN1), 2q34-35 (SLEN2), and 11p15.6 (SLEN3) Genes
Immun.;3 1:S57–S62.
Ramos PS, Brown EE, Kimberly RP, Langefeld CD. (2010). Genetic factors
predisposing to systemic lupus erythematosus and lupus nephritis.
Semin Nephrol.;30(2):164-76.
https://doi:10.1016/j.semnephrol.2010.01.007.
109
The references
Ramos PS, Brown EE, Kimberly RP, Langefeld CD. Genetic factors
predisposing to systemic lupus erythematosus and lupus nephritis.
Semin Nephrol. 2010 Mar;30(2):164-76. doi:
10.1016/j.semnephrol.2010.01.007. PMID: 20347645; PMCID:
PMC2847514.
Rashad NM, El-Shabrawy RM, Said D, El-Shabrawy SM, Emad G. (2019).
Serum Levels of Transforming Growth Factor Beta -1 (TGF-β1) as An
Early No Invasive Marker for Diagnosis of Lupus Nephritis in Systemic
Lupus Erythematosus Patients. Egypt J Immunol.; 26(1):31-42.
Rees F, Doherty M, Grainge MJ, Lanyon P, Zhang W (2017) The worldwide
incidence and prevalence of systemic lupus erythematosus: a systematic
review of epidemiological studies. Rheumatology 56(11):1945–1961
Rekik R, Khanfir MS, Larbi T, Zamali I, et al. (2018). Impaired TGF-β
signaling in patients with active systemic lupus erythematosus is
associated with an overexpression of IL-22. Cytokine. 108:182-189.
http://doi:10.1016/j.cyto.2018.04.011
Rijnink, E. C., Teng, Y. O., Wilhelmus, S., et al. (2017). Clinical and
histopathologic characteristics associated with renal outcomes in lupus
nephritis. Clinical Journal of the American Society of Nephrology,
12(5), 734-743.
Rippon HJ, Bishop AE. (2004). Embryonic stem cells. Cell Prolif. ;37(1):23-34.
https://doi:10.1111/j.1365-2184.2004.00298.x
Rodrigues R. S., Lourenço D. M., Paulo S. L. (2019). Cannabinoid actions on
neural stem cells: implications for pathophysiology. Molecules.;24(7):p.
1350. https://doi:10.3390/molecules24071350
Romito A, Cobellis G. (2016). Pluripotent Stem Cells: Current Understanding
and Future Directions. Stem Cells Int.;2016:9451492.
https://doi:10.1155/2016/9451492
Rosa, S. B., Voltarelli, J. C., Chies, J. A. B., & Pranke, P. (2007). The use of
stem cells for the treatment of autoimmune diseases. Brazilian Journal
of Medical and Biological Research, 40, 1579-1597.
Ruchakorn N, Ngamjanyaporn P, et al. (2019). Performance of cytokine
models in predicting SLE activity. Arthritis Res Ther.,16;21(1):287.
https://doi:10.1186/s13075-019-2029-1
Ruhil S, Kumar V, Rathee P. (2009). Umbilical cord stem cell: an overview.
Curr Pharm Biotechnol. 10(3):327-34.
https://doi:10.2174/138920109787847529
Sabry A, Sheashaa H, El-Husseini A, El-Dahshan K, Abdel-Rahim M,
Elbasyouni SR. Intercellular adhesion molecules in systemic lupus
erythematosus patients with lupus nephritis. Clin Rheumatol. 2007
110
The references
112
The references
113
The references
114
The references
Worku MG. (2021). Pluripotent and Multipotent Stem Cells and Current
Therapeutic Applications: Review. Stem Cells Cloning. 12;14:3-7.
https://doi:10.2147/SCCAA.S304887.
Wszoła M, Nitarska D, Cywoniuk P, Gomółka M, Klak M. (2021). Stem Cells
as a Source of Pancreatic Cells for Production of 3D Bioprinted Bionic
Pancreas in the Treatment of Type 1 Diabetes. Cells. 18;10(6):1544.
https://doi:10.3390/cells10061544.
Wu, X., Jiang, J., Gu, Z. et al. (2020). Mesenchymal stromal cell therapies:
immunomodulatory properties and clinical progress. Stem Cell Res
Ther 11, 345. https://doi.org/10.1186/s13287-020-01855-9
Xie, Min, Cuifang Li, Zhou She, Feifeng Wu, et al. (2022). Human umbilical
cord mesenchymal stem cells derived extracellular vesicles regulate
acquired immune response of lupus mouse in vitro. Scientific Reports
12, no. 1: 13101.
Xu, Jianyong; Chen, Jieting; Li, Wenlei; et al. (2020). Additive Therapeutic
Effects of Mesenchymal Stem Cells and IL-37 for Systemic Lupus
Erythematosus. JASN 31(1):p 54-65,
https://DOI:10.1681/ASN.2019050545
Xu, T., Li, X., Guo, Y., Uhlin, E., Holmberg, L., Mitra, S., ... & Sundström, E.
(2022). Multiple therapeutic effects of human neural stem cells derived
from induced pluripotent stem cells in a rat model of post-traumatic
syringomyelia. EBioMedicine, 77.
Yang C, Sun J, Tian Y, et al. (2021). Immunomodulatory Effect of MSCs and
MSCs- Derived Extracellular Vesicles in Systemic Lupus
Erythematosus, Front. Immunol., 12:3652,
https://DOI=10.3389/fimmu.2021.714832
Yang Q, Liu Y, Chen G, et al. (2021). An Overview of the Safety, Efficiency,
and Signal Pathways of Stem Cell Therapy for Systemic Lupus
Erythematosus. Stem Cells Int. 13;2021:2168595.
https://doi:10.1155/2021/2168595
Yang X, Yang J, Li X, Ma W, Zou H. (2018). Bone marrow-derived
mesenchymal stem cells inhibit T follicular helper cell in lupus-prone
mice. Lupus.;27(1):49-59. https://doi:10.1177/0961203317711013
Ye L, Swingen C, Zhang J. (2013). Induced pluripotent stem cells and their
potential for basic and clinical sciences. Curr Cardiol Rev. 1;9(1):63-72.
https://doi:10.2174/157340313805076278
Yi, P., Xu, X., Qiu, B., & Li, H. (2020). Impact of chitosan membrane culture
on the expression of pro‑and anti‑inflammatory cytokines in
mesenchymal stem cells. Experimental and Therapeutic Medicine,
20(4), 3695-3702.
115
The references
116
The references
117
Annexes
Appendix
(Annexes 1)
BALB/c Mouse Bone Marrow Mesenchymal Stem Cells (BCBMMSCs)
Product Description:
BALB/c Mouse Bone Marrow Mesenchymal Stem Cells are derived from the
tibias of pathogen-free laboratory adult mice. Cells are grown in tissue culture
plates with Cell Biologics’ Culture Medium for 7-15 days. Cultures are then
expanded. Prior to shipping, cells at passage 1 are detached and cryopreserved in
vials. Each vial contains 1x106 cells per ml and is delivered frozen. Cells are
negative for bacteria, yeast, fungi, and mycoplasma. Cells can be expanded for 2 or
3 passages at a split ratio of 1:2 under the cell culture conditions specified by Cell
Biologics. Repeated freezing and thawing of cells is not recommended.
MBMMSCs are tested for expression of markers using antibodies, CD44, Sca-1
and CD29 by flow cytometry.
Laboratory Applications:
MBMMSCs can be used in standard biochemical procedures include PCR,
Western blotting, immunoprecipitation, or cell derivatives for desired research
applications.
Authorized Uses of Cell Biologics Products:
MBMMSCs from Cell Biologics are distributed for internal research purposes
only. Our products are not authorized for human use, for in vitro diagnostic
procedures, or for therapeutic procedures. Transfer or resale of any Cell Biologics’
cells or products from the purchaser to other markets, organizations or individuals
is prohibited by Cell Biologics, without the company’s written consent. Cell
Biologics’ Terms and Conditions must be accepted before submitting an order.
Medium
Review the information provided on the Cell Biologics website about
appropriate culture media. Use pre-warmed (37°C) cell culture media (30-50 ML)
to recover cryo-preserved cells and when changing media or splitting cells.
Handling of Arriving Live Cells:
When you receive the live cells in a T25 or T75 flask, remove the sticker from
the filter cap, and keep the flask with 6-20 ml existing medium in 37°C CO2
incubator for 1 hour before replacing the desired CellBiologics' medium. Either
split the 95-100% confluent cells from a T25 flask to a T75 flask after 1 hour or let
the cells grow in the T25 flask with the desired Medium for 12-48 hours before
subculturing cells. The recommended split ratio for primary cells is 1:2.
Cell Recovery from Cryovial:
• Quickly thaw cells in cryo-vial by incubating them in a 37°C water bath for <1
min until there is just a small bit of ice left in the vial.
• Promptly remove the vial and wipe it down with 70% ethanol.
118
Appendix
• Transfer cells from the vial to a sterile centrifuge tube. Add 8-10 ml of Cell
Biologics’ Medium.
• Flush the vial with an additional 0.5-1 ml of medium to ensure complete transfer
of cells to the centrifuge tube.
• Centrifuge cells at 120 g for 5 minutes.
• Aspirate the supernatant and suspend the cell pellet in 6 ml of Culture Medium.
• Add suspended cells into a flask or plate.
• Place the T25 flask in a humidified, 5% CO2 incubator at 37°C.
• Change culture media the following day to remove non-adherent cells and
replenish nutrients.
• Change cell culture medium every day when cells are >70% confluent.
• Cells should be checked daily under a microscope to verify appropriate cell
morphology.
Expansion of Cultured Primary Cells:
• Remove and discard the cell culture media from the flask.
• Flush the adherent layer 1 time using a 5 ml sterile pipette with sterile PBS (1X)
without calcium and magnesium to dislodge loosely attached cells and remove
fraction.
• Remove and discard the wash solution from the flask.
• Incubate cells with warm (37°C) 0.25% Trypsin-EDTA solution for 2-5 minutes.
Use 3.0 ml of Trypsin-EDTA solution when collecting cells in a T75 flask, and 2
ml when using a T25 flask. As soon as cells have detached (the flask may require a
few firm gentle taps), add 8-10 ml of Cell Culture Medium supplemented with 5-
10 % FBS to a T25 or T75 flask.
• Plate cells in fresh flasks or plates precoated with Gelatin-Based Coating Solution
in a humidified, 5%-CO2 incubator at 37°C.
• Change culture media the following day to remove non-adherent cells and
replenish nutrients.
• Cells should be checked daily under a microscopy to verify appropriate cell
morphology.
• Change culture medium every 24-48 hours. Please note that the medium should
be changed every day when cells are >70% confluent to remove non-adherent cells
and replenish nutrients, pre-wash cells with 1X PBS 1-2 times whenever replacing
the medium.
119
Appendix
(Annexes 2)
Sample Preparation:
1. Serum: Place collected whole blood in refrigerator at 4℃ overnight. Then
centrifuge for 10min at 1000-3000rpm. Take supernatant and either test
immediately or place samples at -20℃/-80℃ (1-3 months) for storage.
2. Plasma: Take EDTA, sodium citrate, or heparin as anticoagulant, and add to
plasma. Mix well. Centrifuge mixture for 10min at 1000-3000rpm. Take
supernatant and test immediately or place samples at -20℃ /-80℃ (1-3 months) for
storage.
3. Tissue homogenate: Take tissue slices and wash them in 0.01M PBS; Add a
tissue protein extraction reagent according to proportion of 1g to 5-10mL and mix
on ice bath. After sufficient homogenization, please then centrifuge for 10min at
5000-10000rpm. Take supernatant for immediate testing, or place samples at -
20℃/-80℃ (1-3 months) for storage.
4. Cell culture: Centrifuge for 10min at 1000-3000rpm. Take supernatant for
immediate testing, or place samples at -20℃/-80℃ (1-3 months) for storage.
5. For urine, ascites, cerebrospinal fluid, etc: Centrifuge for 10min at 1000-3000
rpm. Take supernatant for immediate testing, or place samples at -20℃/-80℃ (1-3
months) for storage.
Test preparation:
121
Appendix
1. Please remove ELISA Kit from refrigerator 20 minutes in advance, and begin
test once it has been brought to room temperature.
2. Dilute the concentrated washing buffer with double distilled water (1:25).
Return unused quantity back to the box.
3. Standard: Add 1.0ml Standard Diluent to lyophilized standard vial and allow
sitting for 30 min. After the standard has completely dissolved, mix it slightly and
mark with a label on the tube. It is recommended to use the following
concentration values for the standard curve: 20, 10, 5, 2.5, 1.25, 0.625,
0.312ng/mL. Note: Make absolutely sure the lyophilized standard completely
dissolved and well mixed.
4. Legend of standard sample dilution method: Take 7 clean tubes and label them
with their expected concentrations (10, 5, 2.5, 1.25, 0.625, 0.312, 0ng/mL). Add
300μL Standard Diluent into each tube. Pipette out 300μl diluent from the
reconstituted standard and add to the tube labeled 10ng/mL and mix well. Further
Pipette out 300μl diluent from the 10ng/mL tube, and add to the 5ng/mL, and mix
well. Repeat these steps through the 0.312ng/mL standard. Standard Diluent in the
0ng/mL tube is the negative control.
Washing method:
1. Automatic plate-washing: The required amount of wash buffer is 350μl, and the
injection and extraction intervals should be ~20-30secs. Please be well aware of
the operation before putting the machine into practice.
2. Manual plate-washing: add 350μl wash buffer to each well and let stand for
30sec. Shake plate to remove as much liquid as possible, and dab the plate with
absorbent paper if necessary. During the plate-washing process, please pay close
attention to the wash buffer-adding steps to avoid contamination and well-jumping.
Step Summary of operating procedures:
1 -Prepare reagents, samples and the standards.
2 -Add the prepared samples and standard & incubate at 37 ℃ for 90 minutes.
3 -Wash 2x, add antigen solution & incubate at 37 ℃ for 60 minutes.
4 -Wash 3x, then add the Enzyme working solution & incubate at 37 ℃ for 30
minutes.
122
Appendix
5 -Wash 5x, then add the Color Reagent solution & incubate at 37 ℃ up to 30
minutes.
6 -Add the Color Reagent C.
7 -Use microplate readers to measure OD within 10 minutes of adding Color
Reagent C.
8 -Calculate the content of samples being tested
123
Appendix
(Annexes 3)
Mouse anti -double –stranded DNA (anti -ds DNA) ELISA Kit
This product is suitable for in vitro quantitative detection of Mouse serum,
plasma or cell culture supernatant and organizations in the natural and recombinant
anti -ds DNA concentration. Detection of other special sample please contacts our
technical support. The kit is for research use only. Please read the instructions
carefully before using and check the kit components. This kit employs Double
Antigen Sandwich Technique. The principle of Double Antigen Sandwich is based
on characteristics of the tested antigen with more than two valances which can
identify coated antigen and detection antigen at same time.
Procedure:
This experiment use double-sandwich Elisa technique and the ELISA Kit
provided is typical. The pre-coated antigen is Mouse anti -ds DNA antigen.
Samples and antigen are added into ELISA plate wells and washed out with PBS
or TBS. Then Avidin-peroxidase conjugates are added to ELISA wells in order;
Use TMB substrate for coloring after reactant thoroughly washed out by PBS or
TBS. TMB turns into blue in peroxidase catalytic and finally turns into yellow
under the action of acid. The color depth and the testing factors in samples are
positively correlated. Schematic diagram of the Mouse anti -ds DNA ELISA kits
the first step The second step
124
Appendix
125
Appendix
4. Legend of standard sample dilution method: Take 7 clean tubes and label them
with ②, ③, ④, ⑤, ⑥, ⑦, ⑧ respectively. Add 300μl standard sample diluent
into each tube. Pipette out 300μl diluent from tube ① to tube ② and mix well.
Further Pipette out 300μl diluent from tube ② to tube ③, and mix well. Repeat
steps above up to tube ⑦. Standard sample dilution in tube⑧is negative control.
5. Mouse anti -ds DNA antigen liquid: Referring to needed amount, employ
antigen diluent to dilute the concentrated antigen (1:100) to form antigen liquid.
The preparation should be done 30 min in advance. And it’s only for use on that
day
6. Enzyme-conjugate liquid: Referring to needed amount, dilute the concentrated
enzyme-conjugate by enzyme-conjugate diluent (1:100) to form enzyme-conjugate
liquid. The preparation should be done 30 min in advance. And it’s only for use on
that day.
7. Color Reagent liquid: Prepare Color Reagent liquid 30 min in advance with
Color Reagent A and Color Reagent B by the proportion of 9:1.
Washing method:
1. Automatic plate-washing machine: The required amount of lotion is 350μl and
the injection and extraction interval should be 20-30secs. Be well aware of the
operation instruction before putting the machine into practice.
2. Manual plate-washing machine: add 350μl lotion to each well and keep it still
for 30secs. Shake individual wells as dry as you can and clean them with absorbent
paper. During the plate-washing process, pay attention to the lotion-adding step to
avoid contamination and well-jumping.
Steps:
1.Take out needed strips from zip lock bag which balances to room temperature.
The unused strips and desiccant should be put back into the sealed aluminum foil
bag at 2-8℃ for storage.
2. Set aside blank wells.
3. Add samples or different concentration of Mouse anti -ds DNA standard
samples to corresponding wells (100μl for each well), 0ng/ml well should be filled
with standard diluent. Seal the reaction wells with adhesive tapes, hatching in
incubator at 37℃ for 90 min.
4. Prepare Mouse anti -ds DNA antigen liquid 30min in advance.
5. Wash the Elisa plate 2 times
6. Add the Mouse anti -ds DNA antigen liquid to each well (100μl for each). Seal
reaction wells with adhesive tapes, hatching in incubator at 37℃ for 60 min.
7. Prepare enzyme-conjugate liquid 30min in advance.
8. Wash the Elisa plate 3 times
126
Appendix
9. Add enzyme-conjugate liquid to each well except blank wells (100μl for each).
Seal the reaction wells with adhesive tapes, hatching in incubator at 37℃ for 30
min.
10. Wash the Elisa plate 5 times.
11. Add 100μl Colour Reagent liquid to individual well, hatching in dark incubator
at 37℃. When color for high concentration of standard curve become darker and
color gradient appears, the hatching can be stopped. The chromogenic reaction
should be controlled within 30 min.
12. Add 100μl Colour Reagent C to individual well. Mix well. Read OD(450nm
)within 10 min.
Result determination:
1. OD value of each sample and specimen should minus that of blank well.
2. Draw standard curve manually. Take concentration value of samples as abscissa
and OD readings as vertical coordinate. Use smooth line to connect each
coordinate point of standard sample. The concentration of samples can be found by
checking sample OD reading. It is recommended to employ the professional curve
software to analyze and compute the result.
3. If the sample OD is higher than the upper limit of standard curve, the sample
should be re-diluted and the experiment rerun. Multiply the result by dilution factor
when calculating the unknown.
Reference curve
127
Appendix
(Annexes 4)
BALB/C model mice:
Also Known As:C, BALBc, BALB, BALB/c BALB/cJ is a commonly used
inbred. Key traits include a susceptibility to developing the demyelinating disease
upon infection with Theiler's murine encephalomyelitis virus. The BALB/cJ
substrain is susceptible to Listeria, all species of Leishmania, and several species
of Trypanosoma, but is resistant to experimental allergic orchitis.
Genetic Background RESEARCH APPLICATIONS:
Immunology, Inflammation and Autoimmunity Research, Cardiovascular
Research, Cancer Research, Neurobiology Research, Hematological Research,
Mouse/Human Gene Homologs.
Detailed Description
BALB/c mice are particularly well known for the production of plasmacytomas
following injection with mineral oil forming the basis for the production of
monoclonal antibodies. Although not all BALB/c substrains have been examined
for plasmacytoma induction, substrains derived from the Andervont (An) lineage
(which includes BALB/cByJ) typically are susceptible, while those descended
from BALB/cJ are resistant.
BALB/c mice immunized with PLP180-199 develop an atypical form of
experimental autoimmune encephalomyelitis in which susceptibility is determined
by location. BALB/c mice are predisposed to dystrophic cardiac calcinosis. Using
BALB/cByJ and BALB/cJ mice obtained from Jackson Laboratory.
Development
This is a substrain of BALB (Bagg’s albino) derived initially from the BALB
strain maintained by MacDowell at Cold Spring Harbor, eventually sent to G.D.
Snell at the University of Texas and brought by Snell to the Jackson Laboratory in
1932 at generation F26. Three branches from Snell’s BALB/c were separated
during the period 1937-1939, Andervont (BALB/cAn), Scott (BALB/cSc), and
Green (BALB/cGn). The lineage from BALB/cSc eventually became BALB/cJ in
1941 at generation F41.
This mouse can be used to support research in many areas including:
Cardiovascular Research, Diet-Induced Atherosclerosis, Cancer Research,
Increased Tumor Incidence, Mammary Gland Tumors, Research Tools,
Monoclonal antibodies, Myeloma, infectious Disease, Neurodevelopmental
Defects, Inflammation, Autoimmunity, Allergic encephalomyelitis (EAE),
Hematological Research, Hemoglobin Defects, and Thalassemia, beta.
128
Appendix
(Annexes 5)
Stability:
This kit is guaranteed to perform as defined if stored and handled as instructed
according to this datasheet and the Certificate of Analysis, which is included with
the reagents. Expiration date is indicated on the box label.
Reagent preparation
• Coating Buffer (1X): Make a 1:10 dilution of PBS (10X) in deionized water.
• Capture Antibody: Dilute capture antibody (250X) 1:250 in Coating Buffer (1X).
• 5X ELISA/ELISPOT Diluent: Dilute Diluent Concentrate (5X) 1:5 in deionized
water.
• Standard: Reconstitute mouse CCL2 standard by addition of distilled water.
Reconstitution volume is stated on the label of the standard vial. Allow the
standard to reconstitute for 10– 30 minutes. Swirl or mix gently to ensure complete
and homogeneous solubilization (concentration of reconstituted standard = 2000
pg/mL).
Mix well prior to making dilutions. The standard has to be used immediately after
reconstitution and cannot be stored.
• Detection Antibody: Dilute detection antibody (250X) 1:250 in ELISA/ELISPOT
Diluent (1X).
129
Appendix
(Annexes 6)
131
Appendix
(Annexes 7)
Mouse ICAM-1 ELISA Kit:
The Mouse ICAM-1 ELISA Kit is a solid-phase sandwich Enzyme-Linked
Immunosorbent Assay (ELISA) designed to detect and quantify the level of mouse
ICAM-1 in serum, plasma, and cell culture media.
Procedural guidelines:
Review the Procedural guidelines and Plate washing directions in the ELISA
Technical Guide at thermofisher.com for details prior to starting the procedure.
Reagents are lot-specific. Do not mix or interchange different reagent lots from
various kit lots.
Prepare 1X Wash Buffer:
1. Allow Wash Buffer Concentrate (20X) to reach room temperature and mix to
dissolve any precipitated salts.
2. Dilute 20 mL of the Wash Buffer Concentrate into 380 mL of deionized or
distilled water. Label as 1X Wash Buffer.
3. Store the concentrate and 1X Wash Buffer in the refrigerator. Use the diluted
buffer within one month.
Prepare diluent:
Assay Diluent D and Assay Diluent B should be diluted 5-fold with deionized or
distilled water before use.
Prepare biotin conjugate:
1. Briefly spin down the biotin conjugate before use.
2. Add 100 μL of 1X Assay Diluent B into the vial to prepare a biotin conjugate
concentrates.
3. Pipette up and down to mix gently (the concentrate can be stored at 4°C for 5
days).
4. The biotin conjugate concentrate should be diluted 80-fold with 1X Assay
Diluent B and used in step 2 of ELISA procedure.
Sample preparation guidelines:
Collect samples in pyrogen/endotoxin-free tubes. Freeze samples after collection
if samples will not be tested immediately. Avoid multiple freeze-thaw cycles of
frozen samples. Thaw completely and mix well prior to analysis. Avoid the use of
hemolyzed or lipemic sera. If large amounts of particulate matter are present in the
sample, centrifuge or filter sample prior to analysis.
Pre-dilute samples:
1X Assay Diluent D should be used for dilution of serum, plasma, and cell
culture supernatant samples. Dilute serum and plasma 50 - 500-fold. Because
conditions may vary, it is recommended that each investigator determine the
optimal dilution to be used for each application.
Dilute standards:
132
Appendix
133
Appendix
Determine the number of 8-well strips required for the assay. Insert the strips in
the frames for use. Re-bag any unused strips and frames, and store at 2 to 8°C for
future use.
1- Bind antigen a. For the standard curve, add 100 μL of standards to the
appropriate wells. For samples, add 100 μL of diluted samples to the wells.
b. Cover wells and incubate for 2.5 hours at room temperature or overnight at 4°C
with gentle shaking.
c. Discard the solution and wash 4 times with 1X Wash Buffer. Wash by filling
each well with Wash Buffer (300 μL) using a multi-channel Pipette or autowasher.
Complete removal of liquid at each step is essential for good performance. After
the last wash, remove any remaining Wash Buffer by aspirating or decanting.
Invert the plate and blot it against clean paper towels.
2- Add biotin conjugate a. Add 100 μL of prepared biotin conjugate to each well.
b. Incubate for 1 hour at room temperature with gentle shaking.
c. Discard the solution. Repeat the wash as in step 3.
3- Add Streptavidin-HRP a. Add 100 μL of prepared Streptavidin-HRP solution to
each well.
b. Incubate for 45 minutes at room temperature with gentle shaking.
c. Discard the solution. Repeat the wash as in step 3.
4- Add TMB substrate
a. Add 100 μL of TMB Substrate to each well. The substrate will begin to turn
blue.
b. Incubate for 30 minutes at room temperature in the dark with gentle shaking.
5- Add stop solution Add 50 μL of Stop Solution to each well. Tap the side of the
plate gently to mix. The solution in the well changes from blue to yellow.
Reading of the standard curve:
1. Read the absorbance at 450 nm. Read the plate within 30 minutes after adding
the Stop Solution.
2. Use curve-fitting software to generate the standard curve. A four parameter
algorithm provides the best standard curve fit. Optimally, the background
absorbance may be subtracted from all data points, including standards, unknowns
and controls, prior to plotting.
3. Read the concentrations for unknown samples and control from the standard
curve. Multiple value(s) obtained for sample(s) by the appropriate factor to correct
for the sample dilution.
Performance characteristics:
Standard curve: These standard curves are for demonstration only. A standard
curve must be run with each assay.
134
Appendix
Linearity of dilution:
The serum, plasma, and cell culture media samples were spiked with
recombinant mouse ICAM-1, serially diluted in sample diluent and evaluated.
Observed values were compared to expected values to calculate percent recovery
and demonstrate the dilution linearity of the assay.
135
Appendix
(Annexes 8)
Mouse IL-6 ELISA Kit
The Invitrogen™ Mouse IL-6 ELISA Kit is a solid-phase sandwich Enzyme-
Linked Immunosorbent Assay. This assay is designed to detect and quantify the
level of mouse IL-6 in mouse serum, plasma, buffered solution, or cell culture
medium. The assay recognizes both natural and recombinant mouse IL-6. IL-6 is a
21–28 kDa glycoprotein composed of 184 amino acids produced by lymphocytes,
monocytes, fibroblasts, keratinocytes, endothelial cells, mesangial cells, astrocytes,
bone marrow stroma, and tumor cell lines. At the protein level, mouse IL-6 shows
42% homology to human IL-6, while mouse and rat are 93% identical. IL-6 plays a
major role in the regulation of cell growth, hematopoiesis, and inflammation. IL-6
induces maturation of B-cells into antibody-secreting plasma cells and it co-
stimulates T-cell growth and cytotoxic T-cell differentiation. IL-6 increases IL-2
receptor production in T-cells and induces production of IL-2. IL-6 is the major
inducer of acute phase reactions in response to inflammation or tissue injury, and
with IL-1b and TNF-a, IL-6 induces synthesis of acute phase proteins by
hepatocytes.
Before you begin:
Do not mix or interchange different reagent lots from various kit lots.
• Review the Procedural guidelines and Plate washing directions in the ELISA
Technical Guide available at thermofisher.com.
• Allow reagents to reach room temperature before use. Mix to dissolve any
precipitated salts. Prepare 1X Wash Buffer
1. Dilute 16 mL of Wash Buffer Concentrate (25X) with 384 mL of deionized or
distilled water. Label as 1X Wash Buffer.
2. Store the concentrate and 1X Wash Buffer in the refrigerator. Use the diluted
buffer within 14 days.
Sample preparation guidelines:
• Refer to the ELISA Technical Guide at thermofisher.com for detailed sample
preparation procedures.
• Collect samples in pyrogen/endotoxin-free tubes.
• Freeze samples after collection if samples will not be tested immediately. Avoid
multiple freeze-thaw cycles of frozen samples. Thaw completely and mix well (do
not vortex) prior to analysis.
• Avoid the use of hemolyzed or lipemic sera. If large amounts of particulate
matter are present in the sample, centrifuge or filter sample prior to analysis.
Pre-dilute samples:
Because conditions may vary, we recommend that each investigator determine
the optimal dilution for each application.
• Perform sample dilutions with Standard Diluent Buffer.
136
Appendix
• For this assay, serum and plasma samples are diluted 1:2 in Standard Diluent
Buffer when added to wells (see "Perform ELISA" step 1a).
1. Reconstitute Ms IL-6 Standard to 10,000 pg/mL with Standard Diluent Buffer.
Refer to the standard vial label for instructions. Swirl or mix gently and allow the
contents to sit for 10 minutes to ensure complete reconstitution. Use the standard
within 1 hour of reconstitution.
2. Add 50 μL Reconstituted Standard to one tube containing 950 μL Standard
Diluent Buffer and mix. Label as 500 pg/mL mouse IL-6.
3. Add 300 μL Standard Diluent Buffer to each of 7 tubes labeled as follows: 250,
125, 62.5, 31.2, 15.6, 7.8, and 0 pg/mL mouse IL-6.
4. Make serial dilutions of the standard as described below in the dilution diagram.
Mix thoroughly between steps.
5. Remaining reconstituted standard should be discarded or frozen in aliquots at –
80°C for further use. Standard can be frozen and thawed one time only without loss
of immunoreactivity.
137
Appendix
138
Appendix
139
Appendix
(Annexes 9)
Mouse IL-10 ELISA Kit:
Product name Mouse IL-10 ELISA Kit, Detection method Colorimetric, Sample
type Cell culture supernatant, Serum, Plasma, Assay type Sandwich (quantitative).
Assay duration multiple steps standard assay:
Species reactivity Reacts with: Mouse Product overview Abcam’s Mouse IL-10
ELISA kit is an in vitro enzyme linked immunosorbent assay for the quantitative
measurement of IL-10 in serum, plasma and cell culture supernatants. This assay
employs an antibody specific for mouse IL-10 coated on a 96-well plate. Standards
and samples are pipetted into the wells and IL-10 present in a sample is bound to
the wells by the immobilized antibody. The wells are washed and biotinylated anti-
mouse IL-10 antibody is added. After washing away unbound biotinylated
antibody, HRP conjugated streptavidin is pipetted to the wells. The wells are again
washed, a TMB substrate solution is added to the wells and color develops in
proportion to the amount of IL-10 bound. The Stop Solution changes the color
from blue to yellow, and the intensity of the color is measured at 450 nm.
Platform Microplate:
Function Inhibits the synthesis of a number of cytokines, including IFN-gamma,
IL-2, IL-3, TNF and GM-CSF produced by activated macrophages and by helper
T-cells. Tissue specificity produced by a variety of cell lines, including T-cells,
macrophages, mast cells and other cell types. Sequence similarities Belongs to the
IL-10 family. Cellular localization Secreted. Quality guaranteed and expert
technical support Replacement or refund for products not performing as stated on
the datasheet Valid for 12 months from date of delivery Response to your inquiry
within 24 hours.
140
Appendix
(Annexes 10)
Mouse Interferon gamma ELISA Kit:
Sample type Milk, Serum, Assay type Sandwich (quantitative), Assay time 3h
45m, Assay duration Multiple steps standard assay Species reactivity Reacts with:
Mouse Product overview Mouse interferon gamma ELISA kit (IFN Gamma) is
designed for the quantitative measurement of IFN gamma in mouse serum,
buffered solutions and cell culture medium. A monoclonal antibody specific for
IFN gamma has been coated onto the wells of the microtiter strips provided.
Samples, including standards of known IFN gamma concentrations, control
specimens or unknowns are pipetted into these wells. During the first incubation,
the standards or samples and a biotinylated monoclonal antibody specific for IFN
gamma are simultaneously incubated. After washing, the enzyme Streptavidin-
HRP, that binds the biotinylated antibody is added, incubated and washed. A TMB
substrate solution is added which acts on the bound enzyme to induce a colored
reaction product. The intensity of this colored product is directly proportional to
the concentration of IFN gamma present in the samples. This kit will recognize
both endogenous and recombinant mouse IFN gamma. Get results in 90 minutes
with Human IFN gamma ELISA Kit from our SimpleStep ELISA range. Platform
Microplate Function Produced by lymphocytes activated by specific antigens or
mitogens. IFN-gamma, in addition to having antiviral activity, has important
immunoregulatory functions. It is a potent activator of macrophages, it has
antiproliferative effects on transformed cells and it can potentiate the antiviral and
antitumor effects of the type I interferons. Tissue specificity Released primarily
from activated T lymphocytes. Involvement in disease In Caucasians, genetic
variation in IFNG is associated with the risk of aplastic anemia (AA). AA is a rare
disease in which the reduction of the circulating blood cells results from damage to
the stem cell pool in bone marrow. In most patients, the stem cell lesion is caused
by an autoimmune attack. T-lymphocytes, activated by an endogenous or
exogenous, and most often unknown antigenic stimulus, secrete cytokines,
including IFN-gamma, which would in turn be able to suppress hematopoiesis.
141
Appendix
(Annexes 11)
Mouse TGF beta 1 ELISA Kit:
Enzyme-linked Immunosorbent Assay for quantitative detection of mouse TGF
beta 1. The mouse TGF beta 1 ELISA is an enzyme-linked immunosorbent assay
for the quantitative detection of mouse TGF beta 1. Transforming growth factor
beta (TGF beta) is a pleiotropic cytokine that exhibits a broad spectrum of
biological and regulatory effects on the cellular and organism level. It plays a
critical role in cellular growth, development, differentiation, proliferation,
extracellular matrix synthesis and degradation, control of mesenchymalepithelial
interactions during embryogenesis, immune modulation, apoptosis, cell cycle
progression, angiogenesis, adhesion and migration and leukocyte chemotaxis. It
has both tumor suppressive and tumor promoting activities and is highly regulated
at all levels.
TGF beta is the first recognized protein of at least 40 of the TGF beta
superfamily of structurally related cytokines. Three isoforms (TGF beta 1-3) have
been described in mammals. (Each isoform is encoded by a unique gene on
different chromosomes. All bind to the same receptors.) They are synthesized by
most cell types and tissues. Cells of the immune system mainly express TGF beta
1. The initially sequestered, inactive LTGF beta (latent TGF beta) requires
activation (cleavage and dissociation of its LAP (latency associated peptide)
region) before it can exert biological activity. LTGF beta can also be bound to LTB
(latent TGF beta binding protein) to form a large latent complex (LLC). TGF beta
forms homodimers, and its subunits of 12.5 kDa each are bound via disulphide
bridges. TGF beta signal transduction is mediated via the TGF beta receptors Type
II and I, phosphorylation and conformational changes.
Sample collection and storage instructions:
Cell culture supernatant, serum and plasma (EDTA, citrate) were tested with this
assay. Other biological samples might be suitable for use in the assay. Remove
serum or plasma from the clot or cells as soon as possible after clotting and
separation.
Preparation of reagents:
Buffer Concentrates should be brought to room temperature and should be
diluted before starting the test procedure. If crystals have formed in the Buffer
Concentrates, warm them gently until they have completely dissolved. Wash buffer
(1x) Pour entire contents (50 mL) of the Wash Buffer Concentrate (20x) into a
clean 1000 mL graduated cylinder. Bring to final volume of 1000 mL with glass-
distilled or deionized water. Mix gently to avoid foaming. Transfer to a clean wash
bottle and store at 2° to 25℃. Please note that Wash Buffer (1x) is stable for 30
days. Wash Buffer (1x) may also be prepared as needed Pour the entire contents (5
mL) of the Assay Buffer Concentrate (20x) into a clean 100 mL graduated
142
Appendix
cylinder. Bring to final volume of 100 mL with distilled water. Mix gently to avoid
foaming. Store at 2° to 8℃. Please note that the Assay Buffer (1x) is stable for 30
days. Assay Buffer (1x) may also be prepared as needed according to the following
table
Mouse TGF beta 1 standard:
1. Reconstitute mouse TGF beta 1 standard by addition of distilled water.
Reconstitution volume is stated on the label of the standard vial. Swirl or mix
gently to insure complete and homogeneous solubilization (concentration of
reconstituted standard = 4 ng/mL). Allow the standard to reconstitute for 10-30
minutes.
2. Mix well prior to making dilutions.
3. After usage remaining standard cannot be stored and has to be discarded.
4. Standard dilutions can be prepared directly on the microwell plate or
alternatively in tubes.
External standard dilution:
1. Label 7 tubes, one for each standard point: S1, S2, S3, S4, S5, S6, S7.
2. Prepare 1:2 serial dilutions for the standard curve as follows: Pipette 225 μL of
Assay Buffer (1x) into each tube.
3. Pipette 225 μL of diluted standard (concentration of standard = 4000 pg/mL)
into the first tube, labelled S1, and mix (concentration of standard 1 = 2 ng/mL).
4. Pipette 225 μL of this dilution into the second tube, labelled S2, and mix
thoroughly before the next transfer.
5. Repeat serial dilutions 5 more times thus creating the points of the standard
curve. Assay Buffer (1x) serves as blank.
Test protocol:
1. Prepare your serum and plasma samples before starting the test procedure.
Dilute serum and plasma samples with Assay Buffer (1x) according to the
following scheme: 20 μL sample + 920 μL Assay Buffer (1x) Add 30 μL 1N HCI
(see “Materials required but not provided“ on page 2) to 940 μL prediluted sample,
mix and incubate for 1 hour at room temperature. Neutralize by addition of 30 μL
1N NaOH
2). Vortex! Prepare your cell culture supernatant samples before starting the test
procedure. Dilute cell culture supernatant samples with Assay Buffer (1x)
according to the following scheme: 20 μL sample + 180 μL Assay Buffer (1x) Add
20 μL 1N HCI to 200 μL prediluted sample, mix and incubate for 1 hour at room
temperature. Neutralize by addition of 20 μL 1N NaOH.
2. Determine the number of microwell strips required to test the desired number of
samples plus appropriate number of wells needed for running blanks and standards.
Each sample, standard, blank and optional control sample should be assayed in
143
Appendix
duplicate. Remove extra microwell strips from holder and store in foil bag with the
desiccant provided at 2°-8℃ sealed tightly.
3. Wash the microwell strips twice with approximately 400 μL Wash Buffer per
well with thorough aspiration of microwell contents between washes. Allow the
Wash Buffer to sit in the wells for about 10 – 15 seconds before aspiration. Take
care not to scratch the surface of the microwells. After the last wash step, empty
wells and tap microwell strips on absorbent pad or paper towel to remove excess
Wash Buffer. Use the microwell strips immediately after washing. Alternatively
microwell strips can be placed upside down on a wet absorbent paper for not
longer than 15 minutes. Do not allow wells to dry.
4. Standard dilution on the microwell plate, Add 100 μL of Assay Buffer (1x) in
duplicate to all standard wells. Pipette 100 μL of prepared standard. Mix the
contents of wells A1 and A2 by repeated aspiration and ejection (concentration of
standard 1, S1 = 2000.0 pg/mL), and transfer 100 μL to wells B1 and B2,
respectively. Take care not to scratch the inner surface of the microwells. Continue
this procedure 5 times, creating two rows of mouse TGF beta 1 standard dilutions
ranging from 2000.0 to 31.3 pg/mL. Discard 100 μL of the contents from the last
microwells (G1, G2) used. In case of an external standard dilution, pipette 100 μL
of these standard dilutions (S1 - S7).
5. Add 100 μL of Assay Buffer (1x) in duplicate to the blank wells.
6. For serum and plasma samples add 80 μL of Assay Buffer (1x) to the sample
wells. For cell culture supernatant samples add 60 μL of Assay Buffer (1x) to the
sample wells. (It is absolutely necessary to vortex the samples!)
7. For serum and plasma samples add 20 μL of each pretreated sample in duplicate
to the sample wells. For cell culture supernatant samples add 40 μL of each
pretreated sample in duplicate to the sample wells. (It is absolutely necessary to
vortex the samples!)
8. Cover with an adhesive film and incubate at room temperature (18 to 25℃) for 2
hours, on a microplate shaker. (Shaking is absolutely necessary for an optimal test
performance.)
9. Prepare Biotin-Conjugate.
10. Remove adhesive film and empty wells. Wash microwell strips 5 times
according to 3 of the test protocol. Proceed immediately to the next step.
12. Cover with an adhesive film and incubate at room temperature (18 to 25℃) for
1 hour, on a microplate shaker. (Shaking is necessary for the test performance.)
13. Prepare Streptavidin-HRP (refer to Preparation of Streptavidin- HRP
“Streptavidin-HRP“ on page 3).
14. Remove adhesive film and empty wells. Wash microwell strips 5 times
according to 3. of the test protocol. Proceed immediately to the next step.
15. Add 100 μL of diluted Streptavidin-HRP to all wells, including the blank wells.
144
Appendix
16. Cover with an adhesive film and incubate at room temperature (18° to 25℃)
for 30 minutes, on a microplate shaker. (Shaking is absolutely necessary for an
optimal test performance.)
17. Remove adhesive film and empty wells. Wash microwell strips 5 times
according to 3 of the test protocol. Proceed immediately to the next step.
18. Pipette 100 μL of TMB Substrate Solution to all wells.
19. Incubate the microwell strips at room temperature (18° to 25℃) for about 30
min. Avoid direct exposure to intense light. The color development on the plate
should be monitored and the substrate reaction stopped before positive wells are no
longer properly recordable. Determination of the ideal time period for color
development has to be done individually for each assay. Adding the stop solution
when the highest standard has developed a dark blue color. Alternatively the color
development can be monitored by the ELISA reader at 620 nm. The substrate
reaction should be stopped as soon as Standard 1 has reached an OD of 0.9 – 0.95.
20. Stop the enzyme reaction by quickly pipetting 100 μL of Stop Solution into
each well. It is important that the Stop Solution is spread quickly and uniformly
throughout the microwells to completely inactivate the enzyme. Results must be
read immediately after the Stop Solution is added or within one hour if the
microwell strips are stored at 2 - 8℃ in the dark.
21. Read absorbance of each microwell on a spectrophotometer using 450 nm as
the primary wave length (optionally 620 nm as the reference wave length; 610 nm
to 650 nm is acceptable). Blank the plate reader according to the manufacturer's
instructions by using the blank wells. Determine the absorbance of both the
samples and the standards.
Calculation of results:
• Calculate the average absorbance values for each set of duplicate standards and
samples. Duplicates should be within 20 percent of the mean value.
• Create a standard curve by plotting the mean absorbance for each standard
concentration on the ordinate against the mouse TGF beta 1 concentration on the
abscissa. Draw a best fit curve through the points of the graph (a 5-parameter curve
fit is recommended).
• To determine the concentration of circulating mouse TGF beta 1 for each sample,
first find the mean absorbance value on the ordinate and extend a horizontal line to
the standard curve. At the point of intersection, extend a vertical line to the
abscissa and read the corresponding mouse TGF beta 1 concentration.
• If instructions in this protocol have been followed, serum and plasma samples
have been diluted 1:250 (20 μL sample + 920 μL Assay Buffer (1x) ( = 1:50) +
30μL 1N HCl + 30μL 1N NaOH and 20 μL pretreated sample + 80 μL Assay
Buffer (1x) ( = 1:5)) and cell culture supernatant samples have been diluted 1:30
(20 μL sample + 180 μL Assay Buffer (1x) + 20μL 1N HCl + 20μL 1N NaOH ( =
145
Appendix
1:12) and 40 μL pretreated sample + 60 μL Assay Buffer (1x) ( = 1:2.5)) and the
concentration read from the standard curve must be multiplied by the dilution
factor (x 250 or 30, respectively).
• Calculation of samples with a concentration exceeding standard 1 may result in
incorrect, low mouse TGF beta 1 levels. Such samples require further external
predilution according to expected mouse TGF beta 1 values with Assay Buffer (1x)
in order to precisely quantitate the actual mouse TGF beta 1 level.
• It is suggested that each testing facility establishes a control sample of known
mouse TGF beta 1 concentration and runs this additional control with each assay.
If the values obtained are not within the expected range of the control, the assay
results may be invalid.
• The curve cannot be used to derive test results. Each laboratory must prepare a
standard curve for each group of microwell strips assayed. Do not use this
standard curve to derive test results. A standard curve must be run for each group
of microwell strips assayed.
9. Prepare Biotin-Conjugate.
10. Empty and wash microwell strips 5 times with Wash Buffer.
11. Add 100 μL Biotin-Conjugate to all wells.
12. Cover microwell strips and incubate 1 hours at room temperature. (Shaking is
necessary for an optimal test performance.)
13. Prepare Streptavidin-HRP.
14. Empty and wash microwell strips 5 times with Wash Buffer.
15. Add 100 μL diluted Streptavidin-HRP to all wells.
16. Cover microwell strips and incubate 30 minutes at room temperature. (Shaking
is necessary for an optimal test performance.)
17. Empty and wash microwell strips 5 times with Wash Buffer.
18. Add 100 μL of TMB Substrate Solution to all wells.
19. Incubate the microwell strips for about 30 minutes at room temperature
20. Add 100 μL Stop Solution to all wells.
21. Blank microwell reader and measure color intensity at 450 nm.
147
Appendix
(Annexes 12)
Mouse VEGF Immunoassay:
Quantikine™ ELISA, This package insert must be read in its entirety before
using this product for research use only. Not for use in diagnostic procedures, for
the quantitative determination of mouse VEGF concentrations in cell culture
supernates, tissue homogenates, serum, and plasma. Vascular endothelial growth
factor is a potent mediator of both angiogenesis and vasculogenesis in the fetus and
in adults. It is a member of the PDGF family that is characterized by the presence
of eight conserved cysteine residues in a cysteine knot structure and the formation
of anti-parallel disulfide-linked dimers. Alternately spliced isoforms of 120, 164
and 188 amino acids have been found in mice, while 121, 145, 165, 183, 189, and
206 aa isoforms have been identified in humans. In humans, VEGF165 appears to
be the most abundant and potent isoform, followed by VEGF121 and VEGF189.
The same pattern may exist in mice. Isoforms other than VEGF120 and VEGF121
contain basic heparin-binding regions and are not freely diffusible. Mouse
VEGF164 shares 97% aa sequence identity with corresponding regions of rat
VEGF. It also shares 89% aa sequence identity with human and porcine VEGF,
88% with bovine VEGF, and 90% with feline, equine, and canine VEGF. VEGF is
expressed in multiple cells and tissues including skeletal and cardiac muscle,
hepatocytes, osteoblasts, neutrophils, macrophages, keratinocytes, brown adipose
tissue, CD34+ stem cells, endothelial cells, fibroblasts, and vascular smooth
muscle cells. Circulating VEGF levels correlate with disease activity in
autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, and systemic
lupus erythematosus. The Quantikine™ Mouse VEGF Immunoassay is a 4.5 hour
solid phase ELISA designed to measure mouse VEGF in cell culture supernates,
tissue homogenates, mouse serum, and plasma. It contains Sf 21-expressed mouse
VEGF and antibodies raised against the recombinant factor. This immunoassay has
been shown to quantitate the recombinant mouse VEGF accurately. Results
obtained using natural mouse VEGF showed dose-response curves that were
parallel to the standard curves obtained using the recombinant Quantikine kit
standards. These results indicate that this kit can be used to determine relative mass
values for natural mouse VEGF.
PRINCIPLE OF THE ASSAY:
This assay employs the quantitative sandwich enzyme immunoassay technique.
A polyclonal antibody specific for mouse VEGF has been pre-coated onto a
microplate. Standards, control, and samples are pipetted into the wells and any
VEGF present is bound by the immobilized antibody. After washing away any
unbound substances, an enzyme-linked polyclonal antibody specific for mouse
VEGF is added to the wells. Following a wash to remove any unbound antibody-
enzyme reagent, a substrate solution is added to the wells. The enzyme reaction
148
Appendix
yields a blue product that turns yellow when the Stop Solution is added. The
intensity of the color measured is in proportion to the amount of VEGF bound in
the initial step. The sample values are then read off the standard curve.
PHARMPAK CONTENTS:
Each PharmPak contains reagents sufficient for the assay of 50 microplates (96
wells/plate). The package inserts supplied are the same as those supplied in the
single kit packs and because of this, a few minor differences related to the number
of reagents and their container sizes should be noted.
• Sufficient material is supplied to perform at least 50 standard curves; reuse of
each vial may be required. The number of vials, and the number of standard curves
obtained per vial will vary with the analyte.
• Wash Buffer 25X Concentrate is bulk packed in 125 mL bottles containing 100
mL.
SAMPLE COLLECTION:
The sample collection and storage conditions listed below are intended as
general guidelines. Sample stability has not been evaluated. Cell Culture
Supernates - Remove particulates by centrifugation and assay immediately or
aliquot and store samples at ≤ -20 °C. Avoid repeated freeze-thaw cycles. Tissue
Homogenates - The preparation of tissue homogenates will vary depending upon
the tissue type. For this assay, heart, kidney, and spleen tissue from three mice and
lung tissue from eight mice were rinsed with 1X PBS to remove excess blood,
homogenized in 20 mL of 1X PBS, and stored overnight at ≤ -20 °C. After two
freeze-thaw cycles were performed to break the cell membranes, the homogenates
were centrifuged for 5 minutes at 5000 x g. Samples can be assayed immediately
or stored at ≤ -20 °C. Avoid repeated freeze-thaw cycles.
Serum - Allow blood samples to clot for 2 hours at room temperature before
centrifuging for 20 minutes at 2000 x g. Remove serum and assay immediately or
aliquot and store samples at ≤ -20 °C. Avoid repeated freeze-thaw cycles.
Plasma - Collect plasma using EDTA or heparin as an anticoagulant. Centrifuge
for 20 minutes at 2000 x g within 30 minutes of collection, assay immediately or
aliquot and store samples at ≤ -20 °C. Avoid repeated freeze-thaw cycles.
SAMPLE PREPARATION:
Use polypropylene tubes. Serum and plasma samples require a 5-fold dilution
into Calibrator Diluent RD5T. A suggested 5-fold dilution is 50 μL of sample +
200 μL of Calibrator Diluent RD5T. Cell culture supernates and tissue
homogenates may require dilution. The dilution factor is dependent upon sample
values.
REAGENT PREPARATION:
Bring all reagents to room temperature before use. Mouse VEGF Control -
Reconstitute the control with 1.0 mL deionized or distilled water. Mix thoroughly.
149
Appendix
Assay the control undiluted. Wash Buffer - If crystals have formed in the
concentrate, warm to room temperature and mix gently until the crystals have
completely dissolved. To prepare enough Wash Buffer for one plate, add 20 mL of
Wash Buffer Concentrate to (480) mL of deionized or distilled water to prepare
500 mL of Wash Buffer. Substrate Solution - Color Reagents A and B should be
mixed together in equal volumes within 15 minutes of use. Protect from light. 100
μL of the resultant mixture is required per well.
Mouse VEGF Standard - Refer to the vial label for reconstitution volume.
Reconstitute the Mouse VEGF Standard with Calibrator Diluent RD5T. Do not
substitute other diluents. This reconstitution produces a stock solution of (500)
pg/mL. Allow the stock solution to sit for a minimum of 5 minutes with gentle
mixing prior to making dilutions. Use polypropylene tubes. Pipette 200 μL of
Calibrator Diluent RD5T into each tube. Use the stock solution to produce a
dilution series (below). Mix each tube thoroughly before the next transfer. The
undiluted Mouse VEGF Standard (500 pg/mL) serves as the high standard.
Calibrator Diluent RD5T serves as the zero standards (0 pg/mL).
ASSAY PROCEDURE:
Bring all reagents and samples to room temperature before use. It is
recommended that all standards, control, and samples be assayed in duplicate.
1. Prepare all reagents, working standards, control, and samples as directed in the
previous sections.
2. Remove excess microplate strips from the plate frame, return them to the foil
pouch containing the desiccant pack, and reseal.
3. Add 50 μL of Assay Diluent RD1N to each well.
4. Add 50 μL of standard, control, or sample to each well. Mix by gently tapping
the plate frame for 1 minute. Cover with the adhesive strip provided. Incubate for 2
hours at room temperature. A plate layout is provided to record standards and
samples assayed.
150
Appendix
5. Aspirate each well and wash, repeating the process four times for a total of five
washes. Wash by filling each well with Wash Buffer (400 μL) using a squirt bottle,
manifold dispenser, or auto washer. Complete removal of liquid at each step is
essential to good performance. After the last wash, remove any remaining Wash
Buffer by aspirating or decanting. Invert the plate and blot it against clean paper
towels.
6. Add 100 μL of Mouse VEGF Conjugate to each well. Cover with a new
adhesive strip. Incubate for 2 hours at room temperature.
7. Repeat the aspiration/wash as in step 5.
8. Add 100 μL of Substrate Solution to each well. Incubate for 30 minutes at room
temperature. Protect from light.
9. Add 100 μL of Stop Solution to each well. Gently tap the plate to ensure
thorough mixing.
10. Determine the optical density of each well within 30 minutes, using a
microplate reader set to 450 nm. If wavelength correction is available, set to 540
nm or 570 nm. If wavelength correction is not available, subtract readings at 540
nm or 570 nm from the readings at 450 nm. This subtraction will correct for optical
imperfections in the plate. Readings made directly at 450 nm without correction
may be higher and less accurate.
CALCULATION OF RESULTS:
Average the duplicate readings for each standard, control, and sample and
subtract the average zero standard optical density (O.D.). Create a standard curve
by reducing the data using computer software capable of generating a four
parameter logistic (4-PL) curve-fit. As an alternative, construct a standard curve by
plotting the mean absorbance for each standard on the y-axis against the
concentration on the x-axis and draw a best fit curve through the points on the
graph. The data may be linearized by plotting the log of the mouse VEGF
concentrations versus the log of the O.D. and the best fit line can be determined by
regression analysis. This procedure will produce an adequate but less precise fit of
the data. If samples have been diluted the concentration must be read from the
standard curve multiplied by the dilution factor.
This standard curve is provided for demonstration only. A standard curve
should be generated for each set of samples assayed.
151
Appendix
SENSITIVITY:
The minimum detectable dose (MDD) of mouse VEGF is typically less than
(3.0) pg/mL. The MDD was determined by adding two standard deviations to the
mean O.D. value of twenty-zero standard replicates and calculating the
corresponding concentration.
LINEARITY:
To assess the linearity of the assay, samples containing and/or spiked with
various concentrations of mouse VEGF in each matrix were diluted with calibrator
diluent and then assayed.
SAMPLE VALUES:
Serum/Plasma - Forty individual mouse serum samples and nine individual
mouse plasma samples were evaluated for detectable levels of mouse VEGF in this
assay. All samples measured less than the lowest mouse VEGF standard, (7.8)
pg/mL. Tissue Homogenates - Tissue homogenates from mouse heart, lung,
kidney, and spleen tissue were assayed for mouse VEGF and measured (111)
pg/mL, 3680 pg/mL, 157 pg/mL, and 30 pg/mL, respectively.
Cell Culture Supernates:
152
Appendix
P388D1 mouse lymphoma cells (1 x 105) cells/mL were cultured for four days
in RPMI supplemented with 5% fetal bovine serum. The cell culture supernate was
assayed for mouse VEGF and measured 7600 pg/mL. Mouse lung conditioned
media (1 lung, 1-2 mm pieces in 10 mL of RPMI supplemented with 10% fetal
bovine serum) was collected after culturing for 5 days. The cell culture supernate
was assayed for mouse VEGF and measured 4660 pg/mL. Mouse heart
conditioned media (1 heart, 1-2 mm pieces in 10 mL of RPMI supplemented with
10% fetal bovine serum) was collected after culturing for 5 days. The cell culture
supernate was assayed for mouse VEGF and measured (378) pg/mL.
SPECIFICITY:
This assay recognizes both the 164 and 120 amino acid residue forms of mouse
VEGF. The factors listed below were prepared at 50 ng/mL in calibrator diluent
and assayed for cross-reactivity. Preparations of the following factors prepared at
50 ng/mL in a mid-range mouse VEGF control were assayed for interference. No
significant cross-reactivity or interference was observed.
153
The published
Papers
Cellular and Molecular Biology
E-ISSN : 1165-158X / P-ISSN : 0145-5680
www.cellmolbiol.org
Immunomodulatory effects of bone marrow-derived Mesenchymal stem cells in a BALB/c
mouse model on induced Systemic lupus erythematosus (SLE)
Ghassan Khudhair Esmael1,2*, Tarek Rebai3, Khalil Gazar Chelab Al-Nailey4
1
Department of Life Sciences, Faculty of Sciences-Sfax, University of Sfax, Tunisia
2
University of Al-Qadisiyah, veterinary medicine college – Iraq
3
Laboratory Histo-embryology and Cytogenetics, Faculty of Medicine of Sfax, University of Sfax, Tunisia
4
University of Al-Qadisiyah, veterinary medicine college – Iraq
Doi: http://dx.doi.org/10.14715/cmb/2022.69.1.4 Copyright: © 2023 by the C.M.B. Association. All rights reserved.
19
Ghassan Khudhair Esmael et al. / Effects of bone marrow-derived Mesenchymal stem cells, 2023, 69(1): 19-24
The study design Source, USA) and ELISA kit anti-double strand DNA
forty BALB/c mice; divided into four groups, each (Anti-dsDNA) (MyBioSource, USA) is used for the detec-
group consisting of ten mice; the first group is adminis- tion of antibodies levels in animals in all the study groups,
trated (ALD DNA) (50 μg/mouse/SC) in three doses at (0, before and after treatment by BM-MSCs and PBS, accor-
14, 28) days to induce SLE in BALB/c mice. The second ding to company directions.
group is administrated (ALD-DNA) (50 μg/mouse/SC),
three doses at (0, 14, 28) days for inducing SLE; after The BALB/c BM-MSCs preparation
onset, the clinical signs of lupus (after 28 days), also, BALB/c- BMMSCs are provided by (Cell biologics
ANA, and anti-dsDNA are examined for final diagnosis Company, USA) from the bone marrow of pathogen-free.
of SLE. Positive lupus in the second group is treated by BALB/c-MSCs could be used in immunosuppressive
BALB/c-MSCs (CellBiologics company, USA) (0.1×106) trials. The cells are thawing water bath (37°C) for (60) se-
cells/for 10g/IV. The third group is administrated BALB/ conds until the disappearance of the ice bit. The provided
c-MSCs (0.1×106) cells/for 10g/IV. The fourth group cells were put in a centrifuge tube with a culture Medium
(control group) administrated BPs only. (8-10) ml for five minutes at 120 rpm. Remove the super-
natant layer then put the cell in a culture Medium (6) ml.
Induction of SLE Add suspended cells into a flask, place the T25 flask in a
After receiving the shipped mice, some of the shipped humidified CO2 (5%), then incubate at (37°C). The media
mice out of the experimental animals (five mice) were are changed daily (for four days) to remove non-adherent
used for the preparation of activated lymphocyte DNA cells and spread the nutrient elements.
(ALD DNA) for induction lupus in mice according to (10),
as following steps: Administration of BALB/c- BM- MSCs
-Preparation of splenocyte The 2nd and 3rd group received BALB/c- BM-MSCs at
removing the BALB/c mice spleens and kept in RPMI- dose (0.1×106) Cells (IV) through the tail vein to decrease
1640 (Gibco, Ireland), then passing the cells several times and prevent the trapping in the lung.
on nylon mesh, then washing by RPMI-1640 then, Add
fetal bovine serum (Gibco, Ireland) (10%) with Mgluta- The used ELISA kits
mine 2m (Sigma, USA), penicillin G and streptomycin The tested cytokines in the study are IL-10, IL-6, CCL-
(100) mg/ml each one to prevent the contamination. Then, 2, TGFβ1, IFNγ, CCL-5, ICAM, and VEGF using ELISA
splenocytes were diluted to (2×106) cells/ml with concana- kits to the company directions as shown in table (1).
valin A (5 ug/ml) for 48 hours (ConA can stimulate mouse
T-cells for giving rise to many folds (11) (12). Statistical Analysis
-DNA extraction: gDNA extracted activated spleno- The results were analyzed by a one-way ANOVA
cytes were treated with proteinase K (Sigma, USA) and and two-way ANOVA tests using SPSS, V27; the USA).
S1 nuclease (TaKaRa, Japan) and then purified by kit (Ge- LSD test was used to determine the significant difference
netech, China). The nanodrop (Thermo Fisher Scientific, between the groups at the significant level (P≥0.05) (13).
USA) is used at (260) nm for determining DNA concen-
trations. Results
-The immunization: the mice were classified into four
groups. Each one consists of ten mice (G1, G2, G3, G4). 1st ANA and Anti dsDNA levels
and 2nd groups were only immunized with activated ALD Our findings showed that the Anti-dsDNA levels be-
DNA (0.2) ml/ SC with PBS and (CFA) (Sigma, USA), fore treatment were (16.98±0.83) ng/ml, (17.29±0.77) ng/
at three doses of ALD DNA, the period between one and ml, (3.10±3.10) ng/ml, and (3.06±5.01) ng/ml. In contrast,
another was two weeks, at (0, 14, 28) day. Before the third Anti-dsDNA levels after treatment were (16.8±0.90) ng/
dose, the animals showed clinical signs of SLE syndrome ml, (2.45±0.52) ng/ml, (3.00±0.30) ng/ml, and (2.98±0.28)
(nose bleeding). ng/ml in the study groups respectively, wherever does not
show significant differences except for the 2nd group which
ANA and anti-dsDNA Examination showed significant differences between before and after
ELISA kit antinuclear antibodies (ANA) (MyBio- treatment by BM-MSCs at (P<0.05).
20
Ghassan Khudhair Esmael et al. / Effects of bone marrow-derived Mesenchymal stem cells, 2023, 69(1): 19-24
Table 2. level of ANA and Anti dsDNA in serum before and after treatment by BM-MSCs.
Anti dsDNA ng/ml ANA ng/ml
Before treatment by After treatment by Before treatment by After treatment by
BM-MSCs BM-MSCs BM-MSCs BM-MSCs
G1 16.98 ±0.83Aa 16.8±0.90Aa 18.15±1.45Aa 18.07±1.09Aa
G2 17.29 ±0.77Aa 2.45 ±0.52Bb 17.55±0.83Aa 2.71±0.61Bb
G3 3.10±3.10Ba 3.00±0.30Bb 3.70±4.1Ba 3.22±0.64Bb
G4 3.06±5.01Ba
2.98±0.28Bb 3.63±4.87 Ba
3.30±0.75Bb
0.584 0.861
23
Ghassan Khudhair Esmael et al. / Effects of bone marrow-derived Mesenchymal stem cells, 2023, 69(1): 19-24
dulatory capability and clinical potential in immune diseases, J. overexpression of IL-22. Cytokine. 2018 Aug; 108:182-189.
Cell. Immunother., 2016; 2 (1), 3-20, https://doi.org/10.1016/j. http://doi:10.1016/j.cyto.2018.04.011
jocit.2014.12.001 28. Davis LS, Hutcheson J, and Mohan C. The role of cytokines in the
19. Lu R, Munroe ME, Guthridge JM, Bean KM, Fife DA, Chen H, pathogenesis and treatment of systemic lupus erythematosus. J.
Slight-Webb SR, Keith MP, Harley JB, James JA. Dysregulation Interferon Cytokine Res. 2011 Oct; 31(10): 781-789.
of innate and adaptive serum mediators precedes systemic lupus 29. Ohl K, Tenbrock K. Inflammatory cytokines in systemic lupus
erythematosus classification and improves prognostic accuracy erythematosus. J Biomed Biotechnol. 2011 Oct; 16:2011:432595.
of autoantibodies. J. Autoimmun. 2016 Nov; 74:182-193. https:// https://doi:10.1155/2011/432595
doi:10.1016/j.jaut.2016.06.001 30. Ruchakorn, N, Ngamjanyaporn, P, Suangtamai, T, Kafaksom T,
20. Bai, Y, Tong, Y, Liu, Y, & Hu, H. Self-dsDNA in the pathogenesis Polpanumas C, Petpisit V, Pisitkun T, Pisitkun P. Performance of
of systemic lupus erythematosus. Clin. Exp. Immunol., 2018 Jan; cytokine models in predicting SLE activity. Arthritis Res. Ther.
191(1), 1–10. https://doi.org/10.1111/cei.13041 2019 Dec; 21, 1-1. https://doi.org/10.1186/s13075-019-2029-1
21. Sohrabian, A., Parodis, I, Carlströmer-Berthén, N, Frodlund, M, 31. Idborg, H, & Oke, V. Cytokines as Biomarkers in Systemic Lu-
Jönsen, A, Zickert, A, Sjöwall, C, Bengtsson, A, Gunnarsson, I, & pus Erythematosus: Value for Diagnosis and Drug Therapy. Int.
Rönnelid, J. Increased levels of anti-dsDNA antibodies in immune J. Mol. Sci. 2021 Jan; 22(21), 11327. https://doi.org/10.3390/
complexes before treatment with belimumab are associated with ijms222111327
clinical response in patients with systemic lupus erythematosus. 32. Poole, BD, Niewold, TB, Tsokos, GC, & Via, CS. Cytokines in
Arthritis Res. Ther., 2019 Dec; 21(1), 259. https://doi.org/10.1186/ systemic lupus erythematosus. J. biotechnol. biomed., 2010 Oct,
s13075-019-2056-y 735169. https://doi.org/10.1155/2010/735169
22. Godsell, J, Rudloff, I, Kandane-Rathnayake, R, Hoi, A, Nold, MF, 33. Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchy-
Morand, EF, & Harris, J. Clinical associations of IL-10 and IL-37 mal Stem Cell Therapy: Hope for Patients With Systemic Lupus
in systemic lupus erythematosus. Sci. Rep., 2016 Oct; 6, 34604. Erythematosus, Front. Immunol., 2021 Sep, 12: 4062, https://
https://doi.org/10.1038/srep34604 DOI=10.3389/fimmu.2021.728190
23. Yuan, W., DiMartino, SJ, Redecha, PB, Ivashkiv, LB, & Sal- 34. Yang C, Sun J, Tian Y, Li H, Zhang L, Yang J, Wang J, Zhang
mon, JE. Systemic lupus erythematosus monocytes are less res- J, Yan S, Xu D. Immunomodulatory Effect of MSCs and MSCs-
ponsive to interleukin-10 in the presence of immune complexes. Derived Extracellular Vesicles in Systemic Lupus Erythemato-
Arthritis and rheumatism, 2011 Jan; 63(1), 212–218. https://doi. sus, Front. Immunol., 2021; 12:3652, https://DOI=10.3389/
org/10.1002/art.30083 fimmu.2021.714832
24. Waterman, RS, Morgenweck, J, Nossaman, BD, Scandurro, AE, 35. Li, W, Chen, W, Sun, L. An update for mesenchymal stem cell
Scandurro, SA, & Betancourt, AM. Antiinflammatory mesenchy- therapy in lupus nephritis. Kidney Dis., 2021;7(2), 79-89. https://
mal stem cells (MSC2) attenuate symptoms of painful diabetic doi:10.1159/000513741
peripheral neuropathy. Stem Cells Transl. Med., 2012 Jan; 1(7), 36. Zhou, T, Liao, C, Li, HY. Efficacy of mesenchymal stem cells in
557–565. https://doi.org/10.5966/sctm.2012-0025 animal models of lupus nephritis: a meta-analysis. Stem Cell Res.
25. Wang, JH, Liu, XL, Sun, JM, Yang, JH, Xu, DH, & Yan, SS. Role Ther. 2020 Dec; 11(1) 1-10. https://doi.org/10.1186/s13287-019-
of mesenchymal stem cell-derived extracellular vesicles in au- 1538-9
toimmunity: A systematic review. World J Stem Cells, 2020 Aug; 37. Dandan W, Lingyun S, Xiao-Dong C. Chapter 7 - Systemic Lupus
12(8), 879–896. https://doi.org/10.4252/wjsc.v12.i8.879 Erythematosus, A Roadmap to Non-Hematopoietic Stem Cell-
26. Rashad NM, El-Shabrawy RM, Said D, El-Shabrawy SM, Emad based Ther., Academic Press, 2019; 143-172.
G. Serum Levels of Transforming Growth Factor Beta -1 (TGF-β1) 38. Zhou T, Li HY, Liao C, Lin W, Lin S, Clinical Efficacy and
as An Early No Invasive Marker for Diagnosis of Lupus Nephri- Safety of Mesenchymal Stem Cells for Systemic Lupus Erythe-
tis in Systemic Lupus Erythematosus Patients. Egypt J Immunol.; matosus", Stem Cells Int., 2020 Oct; (11): 2020. https://doi.
2019 Jan; 26(1):31-42. PMID: 31332994. org/10.1155/2020/6518508
27. Rekik R, Khanfir MS, Larbi T, Zamali I, Beldi-Ferchiou A, Kam- 39. Bukulmez H, Kumar G. Clinical Use of Mesenchymal Stem Cells
moun O, Marzouki S, Hamzaoui S, Mrad S, Barbouche MR, Hou- in Treatment of Systemic Lupus Erythematosus, Lupus-Need
man MH, Ben Ahmed M. Impaired TGF-β signaling in patients to Know, 2021 May 22; IntechOpen, https://doi:10.5772/inte-
with active systemic lupus erythematosus is associated with an chopen.97261
24
Volume 21 | June 2023 ISSN: 2795-7624
PBS. After finishing the induction and treatment time, all the animals activated for take
the serum for estimation the gene expression of the some of the cytokines and
chemokines genes by using RT-PCR techniques in all the study groups. The findings are
included that the first group showed significant high expression of IL-6, CCL-5/RANTES,
VEGF, ICAM, CCL-2, and IFNγ-encoding genes; and low expression of IL-10 and TGFβ1-
encoding gene as compared with the control group. The second group showed a
significant low expression of IL-6, CCL-5/RANTES, VEGF, ICAM, CCL-2/MCP-1, and IFNγ-
encoding genes while showing significant high expression of IL-10 and TGFβ1-encoding
gene as compared with the control group. The third group doesn't show any changes in
the studied cytokines and chemokines gene expression compared to the control group.
BM-MSCs can effect on the gene expression of many cytokines and chemokine encoding
genes, which treats induced lupus in mice.
Keywords:
Mesenchymal stem cells, gene expression, cytokines genes,
BALB/c mice, lupus
Introduction: lupus is a chronic immune MSCs can regulate the immunological
disorder that affects humans and animals. response in vivo and in vitro in humans and
Mesenchymal stem cells (MSCs) are adult stem mice by two mechanisms: forming of the
cells; they can differentiate into several cell soluble factors (IL-10, PGE2, and TGF-β1); and
type lineages (1). MSCs have by cellular communication (3) (4). T helper
immunomodulatory impacts because they cells are regulating the immune responses by
inhibit Th17 cells and stimuli Treg cells in producing cytokines (5). MSCs have an
autoimmune diseases (2).
Total RNA Extraction and Reverse The fold change are calculated as
Transcriptase: 2-ΔΔCTΔΔCT =ΔCT
RNA was extracted from animals serum ΔCT-ΔCT =CT gene-CT House Keeping gene.
directly by using RNA Extraction Kit Statistical Analysis: The results were
(Addbio/Korea), 3). Extracted RNA was analyzed by an ANOVA test (SPSS, V20; USA).
converted to cDNA by SYBR Green Master Using of LSD test to determine the difference
(Quantabio/Germany). The final volume 25 μl among the groups at P= 0.05 (24).
that consist of (SYBR Green Master Mix 12.5 μl,
F primer and R primer 1 μl, DNA free water 6.5 Results:
μl, RNA template 3 μl and qScript One- Step The gene expression of the IL-10 gene in G1
RT3 1 μl). The total reaction mixture was was low than in control. No significant
incubated into the thermocycler. cDNA differences exist among other groups in gene
synthesis (49) ºC for (10) minutes., Taq expression of the IL-10 gene, as shown in table
activation 95ºC for (2) minutes., PCR cycling (2) and figure (1).
(38 cycles, 95 ºC for (4) seconds, and 59 ºC for
(40) seconds)
Table (2): Values of the fold change of the IL-10 gene
Group Fold change
G1 0.223 B
G2 0.905 A
G3 0.998 A
G4 1A
The same letters haven't significant differences; the different letters have significant
differences
Fold change
1
0.8 G1
0.6 G2
0.4 G3
0.2 G4
0
G1 G2 G3 G4
Figure (1): the chart shows the fold changes of the IL-10 gene
Fold change
2
G1
1.5
G2
1
G3
0.5 G4
0
G1 G2 G3 G4
Figure (2): chart shows the fold changes of the IL-6 gene
The findings showed that the TGF-β1 expression gene was reduced in G1 than in the control. There
are no marked differences among other groups in the TGF-β1 expression gene, as shown in table (4)
and figure (3).
Table (4): The fold change values of the TGF-β1 gene
Group Fold change
G1 0.365 A
G2 0.996 B
G3 0.997 B
G4 1B
The same letters haven't significant differences; the different letters have significant
differences
Fold change
1
0.8 G1
0.6 G2
0.4 G3
0.2 G4
0
G1 G2 G3 G4
Figure (3): chart shows the fold changes of the TGF-β1 gene
Fold change
4 G1
3 G2
2
G3
1
G4
0
G1 G2 G3 G4
Figure (4): chart shows the fold changes of the CCL-2/MCP-1 gene
The current study showed that the gene expression of the CCL-5/RANTES-encoding gene was
increased in G1 than in the control. There are no marked differences among other groups in gene
expression of the CCL-5/RANTES-encoding gene, as shown in table (6) and figure (5).
Table (6): Values of the fold change of CCL-5/RANTES encoding gene
Group Fold change
G1 5.48684 A
G2 0.96846 B
G3 1.00919 B
G4 1B
The same letters haven't significant differences; the different letters have significant
differences
Fold change
6 G1
4 G2
2 G3
G4
0
G1 G2 G3 G4
Figure (5): chart shows the fold changes of the CCL-5 gene
The gene expression of the VEGF-A encoding gene was increased in G1 than in the control. There are
no marked differences among other groups in gene expression of the VEGF-A-encoding gene, as
shown in table (7) and figure (6).
Fold change
5
G1
4
3 G2
2 G3
1 G4
0
G1 G2 G3 G4
Figure (6): chart shows the fold changes of the VEGF-A gene
The current study showed that the ICAM-1 encoding gene expression was increased in G1 than in the
control. There are no marked differences among other groups in gene expression of the ICAM-1-
encoding gene, as shown in table (8) and figure (7).
Table (8): Values of the fold change of the ICAM-1 gene in study groups
Group Fold change
G1 1.34562 A
G2 1.00755 B
G3 1.000888 B
G4 1B
The same letters haven't significant differences; the different letters have significant
differences
Fold change
1.5
G1
1
G2
G3
0.5
G4
0
G1 G2 G3 G4
Figure (7): chart shows the fold changes of ICAM-1 gene in study groups
The current study showed that gene expression of the IFN-γ-encoding gene was increased in G1
than in the control. There are no marked differences among other groups in gene expression of the
IFN-γ encoding gene, as shown in table (9) and figure (8).
Fold change
2.5
G1
2
1.5 G2
1 G3
0.5 G4
0
G1 G2 G3 G4
Figure (8): chart showed the fold changes of the IFN-γ gene in study groups