60

Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

Embed Size (px)

Citation preview

Page 1: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis
Page 2: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis
Page 3: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

Index

Resumen ......................................................................................................................................................... 5

Abstract ........................................................................................................................................................... 7

1. Introduction ..................................................................................................................... 8

1.1 Cancer ................................................................................................................................................... 8

1.2 Pancreatic ductal adenocarcinoma (PDAC) ............................................................................. 9

1.3 The lipid membrane ....................................................................................................................... 10

1.3 Autophagy ......................................................................................................................................... 14

1.4 Lipid membrane alterations in cancer ..................................................................................... 18

1.5 Membrane-Lipid Therapy ............................................................................................................ 19

1.6 Objects of study: Mimetic triglycerides (TGMs) .................................................................. 20

2. Objectives ................................................................................................................................................ 22

3. Materials and methods..................................................................................................................... 23

3.1 Cell lines ............................................................................................................................................. 23

3.2 Cell culture ......................................................................................................................................... 23

3.2.1 Cell lines and growth conditions....................................................................................... 23

3.2.2 Thawing ...................................................................................................................................... 23

3.2.3 Cell passaging / maintaining .............................................................................................. 23

3.2.4. Freezing ..................................................................................................................................... 24

3.2.5 Cellular treatments ................................................................................................................. 24

3.3 Cell proliferation / cytotoxicity studies ................................................................................... 24

3.4 Cell cycle studies ............................................................................................................................. 25

3.5 Western Blot studies ...................................................................................................................... 26

3.5.1 Cell lysate ................................................................................................................................... 26

3.5.2 Protein quantification ............................................................................................................ 27

3.5.3 Western Blot ............................................................................................................................. 27

3.6 Thin Layer Chromatography ....................................................................................................... 29

3.6.1 Lipid extraction ........................................................................................................................ 29

Page 4: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

3.6.2 Thin layer chromatography ................................................................................................. 30

3.7 In vivo studies ................................................................................................................................... 30

3.7.1 Experimental animals............................................................................................................. 30

3.7.2 Determination of the toxicity of the compounds TGM4 and TGM5. .................. 31

3.7.3 Antitumor studies ................................................................................................................... 31

3.8 Data analysis ..................................................................................................................................... 32

4. Results ....................................................................................................................................................... 33

4.1 Mimetic triglycerides (TGMS) inhibit cell viability and proliferation. .......................... 33

4.2 Determination of a safe dose to perform in vivo studies with TGM4 and TGM5... 36

4.3 TGM4 effect on the progression of Mia-PaCa-2 cell line xenograft in nude

immunodepressed mice ...................................................................................................................... 38

4.4 Effect of the TGM4 on the cell cycle. ....................................................................................... 40

4.5 Finding the type of cell death and related proteins behind the TGM4 effect. ........ 43

4.6 Lipid alterations produced by the treatment with TGM4 ................................................ 45

5. Discussion ............................................................................................................................................... 47

5.1 The TGMs, a novel library of compounds, are effective antiproliferative drugs. .... 47

5.2 The TGM4 is an effective antitumor drug, tested on Mia-PaCa-2 xenograft in vivo

models of pancreatic cancer. ............................................................................................................. 48

5.3 TGM4 produces an antiproliferative effect and death by autophagy in cancer cells

PANC-1. ..................................................................................................................................................... 50

6. Conclusions ............................................................................................................................................ 54

7. References ...................................................................................................................... 55

Page 5: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

5

Resumen

El cáncer de páncreas de células ductales (PDAC en inglés) es la cuarta causa de muerte

por cáncer en EEUU y la sexta en Europa. Su difícil diagnóstico precoz, acompañado de

su alto índice de metástasis, hace de este cáncer una enfermedad fatal: la media de

supervivencia desde el diagnóstico es de solo 4-6 meses. La única terapia curativa es la

resección quirúrgica, y fuera de ella no hay tratamientos eficaces disponibles a día de

hoy. El cáncer de páncreas ofrece un escenario donde el desarrollo de nuevas terapias es

necesario, y este trabajo de fin de master estudia el uso de nuevos fármacos de diseño

que funcionan a través la Terapia Lipídica de Membrana como tratamiento contra el

cáncer pancreático.

En este trabajo se realizó un screening del efecto antitumoral in vitro de una nueva

librería de compuestos, los triglicéridos miméticos o TGMs. Estos compuestos

presentaron, en mayor o menor medida, efectos antiproliferativos destacables en una

línea celular de cáncer pancreático.

Se seleccionaron los compuestos TGM4 y TGM5 para realizar estudios in vivo utilizando

un modelo de inducción de tumores xenográficos en ratones inmunodeprimidos NUDE,

mediante la inyección subcutánea de células de cáncer de páncreas Mia-PaCa-2. Ambos

demostraron no ser tóxicos a las dosis estudiadas y ambos mostraron actividad

antitumoral provocando una reducción de la progresión y crecimiento de los tumores

inducidos respecto a los controles. Sin embargo, la eficacia del TGM4 fue mayor a la del

TGM5 y muy similar a la de la Gemcitabina, el fármaco de referencia para el tratamiento

del cáncer de páncreas en humanos. Por los resultados obtenidos, se seleccionó el TGM4

como compuesto del que determinar el mecanismo de acción.

En la investigación del mecanismo de acción del TGM4 se utilizaron células tumorales

ductales PANC-1. Se pudo determinar por citometría de flujo que el TGM4 producía

muerte celular y una disminución del porcentaje de células en fase G0/G1, datos

apoyados por la disminución de la proteína reguladora del ciclo celular Ciclina D3.

Estudios a nivel de proteína revelaron que el TGM4 produce la muerte celular a través de

un estrés reticular (indicado por la sobreexpresión de BIP) que desencadena a través de

CHOP la muerte por autofagia. La formación de autofagosomas fue corroborada por la

aparición de LC3B-II. También se observaron cambios en vías de señalización implicadas

en oncogénesis como son la vía de las MAPK y PI3K/AKT y en el proto-oncogen c-jun.

Finalmente, mediante cromatografía de capa fina se estudió como afectaba el TGM4 a la

composición de la membrana lipídica, revelando cambios en los fosfolípidos cardiolipina

y fosfatidilcolina.

En resumen, los resultados indican que los compuestos incluidos en la librería de TGMs

estudiada tienen efectos antiproliferativos en células tumorales. El TGM4 y TGM5 han

demostrado mantener el efecto en modelos in vivo, con resultados significativos en el

Page 6: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

6

caso del TGM4. Tras los estudios realizados, se puede decir que el TGM4 es una molécula

que por su efecto antiproliferativo a través de la inducción de la autofagia, y por su

aparente inocuidad y efectividad antitumoral en modelos in vivo, podría ser un fármaco

alternativo en el tratamiento del cáncer de páncreas.

Page 7: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

7

Abstract

Pancreatic cancer ductal cells (PDAC) is the fourth leading cause of cancer death in the

US and sixth in Europe. Its difficult early diagnosis accompanied by its high rate of

metastasis makes this a fatal disease: the median survival time from diagnosis is only 4-

6 months. The only curative therapy is surgical resection, and outside there are no

effective treatments available today. Pancreatic cancer requires the development of new

therapies, and this master thesis studies the use of novel synthetic lipidic drugs included

in the Membrane-Lipid Therapy as a treatment for pancreatic cancer.

In this work an in vitro screening to determine the antitumor effect of novel mimetic

triglycerides or TGMs library was performed. These compounds showed a greater or

lesser remarkable antiproliferative effects on a pancreatic cancer cell line.

The compounds TGM4 and TGM5 were selected for in vivo studies in xenograft tumor

models induced by pancreatic cancer cells, Mia-PaCa-2, injected subcutaneously in

immunodepressed nude mice. Both demonstrated to be non-toxic at the tested doses

and showed antitumor activity impairing the tumor growth and progression in

comparison to control. However, TGM4 showed greater efficacy than TGM5, being very

similar to gemcitabine, the standard of care drug for treating pancreatic cancer in

humans. From these results, TGM4 was selected to determine its mechanism of action.

Investigating the mechanism of action of TGM4, pancreatic ductal tumor cells PANC-1

were used. TGM4 produced cell death and decreased the percentage of cells in G0/G1

phase, as determined by flow cytometry. This result was supported by the decrease in

cell cycle regulatory protein cyclin D3. Furthermore, TGM4 provokes cell death through

a reticular stress (indicated by BIP overexpression) that triggers autophagy via CHOP. The

autophagosomes formation was corroborated by the LC3B-II protein modification.

Changes were also observed in signaling pathways involved in oncogenesis, such as the

MAPK and PI3K / AKT pathways and the proto-oncogene c-jun. Finally, Thin-Layer

Chromatography was use to study how TGM4 affected to the composition of the lipid

membrane, showing changes in the phospholipids cardiolipin and phosphatidylcholine.

In summary, the results indicate that the compounds included in the TGMs library have

antiproliferative effect on tumor cells. In fact, TGM4 and TGM5 have shown to be

antitumor drugs on in vivo models, with outstanding results in the case of TGM4. As

conclusion, TGM4 is a molecule which due to its antiproliferative effects through the

induction of autophagy, apparent safety and antitumor effectiveness in vivo models

could be an alternative drug in the treatment of pancreatic cancer.

Page 8: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

8

1. Introduction

1.1 Cancer

Cancer is the name given to a collection of diseases which share a common feature: a

transformation of cells leading to an abnormal and uncontrolled proliferation. Usually,

human cells grow and divide to form new cells as the body needs them, but if the new

cells are mutated or damaged, they die and healthy cells take their place, in cancer this

basic process is altered.

The uncontrolled proliferation leads to the formation of new cells when they are not

needed. Cancer cells differ from normal cells in many ways that allow them to grow out

of control and become invasive. These cells dividing without stopping may form a cell

mass commonly known as tumor, finally able to spread and invade subjacent tissues and

form new tumors in the process known as metastasis.

Figure 1. This illustration encompasses the six hallmark capabilities of tumor cells. From: Hanahan

& Weinberg, 2011.

The transformation of the cells from normal cells to tumor cells is mainly caused by the

sequential acquisition of mutations, which give to the cells the characteristics to grow

and evade the immune and homeostatic barriers (Hanahan and Weinberg, 2000). These

specific properties (Figure 1) are: immune evasion, redirection of metabolic energy,

invasiveness and metastasis, sustained angiogenesis, limitless replicative potential,

evasion of apoptosis, insensitivity to antiproliferative signals and self-sufficiency in

growth signals (Hanahan and Weinberg, 2011). The origin of these changes are extremely

Page 9: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

9

varied, ranging from endogenous factors such as DNA replication errors, loss of

heterozygosity, free radicals generated by metabolism, spontaneous mutations or

congenital genetic predisposition, but they can be also exogenous epigenetic factors

such as ionizing radiation, ultraviolet radiation or abundantly present carcinogens in the

current lifestyle.

Added to the internal nature of this kind of disease, the cells in a tumor are not exactly

equal, being possible to find, in a unique tumor, cells with highly different biochemical,

morphological and immunological features. In addition, cancer cells are able to ignore

signals that normally tell cells to stop dividing, as programmed cell death, or apoptosis,

which the organism uses to get rid of unneeded cells. Cancer cells may be able to

influence the normal cells, molecules, and blood vessels that surround and feed a

tumor—an area known as the microenvironment.

The genetic changes that contribute to cancer affect three main types of genes—proto-

oncogenes, tumor suppressor genes, and DNA repair genes. These changes are

sometimes called “drivers” of cancer.

In that scenario, proto-oncogenes are involved in normal cell growth and division.

However, when these genes are altered, usually being more active than normal, they may

become cancer-causing genes (or oncogenes), allowing cells to grow and survive when

they should not. On the other hand, tumor suppressor genes are also involved in

controlling and stopping cell growth and division. Cells with certain alterations in tumor

suppressor genes may grow in an uncontrolled manner. Finally, DNA repair genes are

involved in fixing damaged DNA, for example the mutations in proto-oncogenes or

tumor suppressor genes. Cells with mutations in DNA repair genes tend to develop

accumulate mutations in several genes. Together, these mutations may cause the cells to

become cancerous

As this introduction has tried to manifest, cancer is a highly complex disease, moreover,

it figure among the leading causes of morbidity and mortality worldwide, with

approximately 14 million new cases and 8.2 million cancer related deaths in 2012 (World

Cancer Report, 2014). The number of new cases is expected to rise by about 70% over

the next 2 decades.

1.2 Pancreatic ductal adenocarcinoma (PDAC)

The pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death

in the United States and sixth in Europe, despite of the relative short incidence of 10-

12:100.000 (Jemal et al. 2009). Every year more than 40.000 people are diagnosed with

Page 10: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

10

PDAC only in the United States, and more than 36.000 die every year due to this fatal

disease.

The clearest risk factor is the advanced age, but now it is known that smoking, diabetes,

obesity, or chronic inflammation of the pancreas, known as pancreatitis, are risk factors

to develop this cancer (Everhart & Wright 1995; Gapstur et al. 2000; Michaud et al. 2001;

de Gonzalez et al. 2003; Stolzenberg-Solomon et al. 2005)

The prognosis for those diagnosed with PDAC is fatal in the vast majority of cases. Fewer

than 5% of all PDAC patients are still alive 5 years after initial diagnosis (Yang et al. 2013).

The median survival time for PDAC patients is only 4-6 months from initial diagnosis.

Unfortunately, surgical resection is still the only potentially curative treatment (Riall et al.

2005) and chemotherapy or radiotherapy are used generally as palliative treatments.

The vast majority of pancreatic cancer patients are today primarily treated with a

palliative intent to reduce symptoms as well as to prolong life for some patients.

Gemcitabine is generally advised as the standard first-line treatment for pancreatic

cancer patients (Burris Ha et al. 1997). Gemcitabine is adeoxycytidine analogue that must

be phosphorylated to become active (gemcitabine diphosphate and gemcitabine

triphosphate). When activated, gemcitabine diphosphate inhibits ribonucleotide

reductase and reduces the intracellular pool of deoxynucleotide triphosphate required

for DNA synthesis.

Despite the bad prognosis, several hallmarks of the biology of this cancer are now

understood, and with unmet clinical needs new treatments are necessary to improve the

prognosis of this fatal disease.

1.3 The lipid membrane

It took nearly two hundred years after the development of the cell theory before a

complete cell membrane theory was developed to explain what separates the cells from

the outside world, but by the 19th century it was accepted that some form of semi-

permeable barrier must exist around a cell.

The composition of that membrane was correctly intuited by Quincke, who in a series of

elegant experiments noted that a cell generally forms a spherical shape in water and,

when broken in half, forms two smaller spheres, as the oil do. The idea of a semi-

permeable membrane means that the membrane is permeable to solvent but

impermeable to solute molecules.

Page 11: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

11

The fluid mosaic model, developed by S. J. Singer and G. L. Nicolson in 1972, consider

the membrane as a lipid bilayer, formed by several different lipids able to move through

the membrane, and where a large quantity of proteins are embedded. The membrane is

not a simple structure with the only function of separate the cell content from the outside,

it is a highly complex structure that allows the cell to communicate with his surround,

and that is the base for the multicellular life. Notwithstanding, the fluid mosaic model is

simplistic and incomplete.

An adequate view of the membrane based on the knowledge achieved in the last years

is that one proposed by Engelman (Engelman, 2005) (Figure 2), a membrane where

protein density is remarkable, where the lipids are heterogeneous and where the relation

between different types of lipids (1400 in average) and thousands of proteins is intensive.

Figure 2. A good perspective of the complex fluid mosaic model, where different proteins and

lipids are components of the lipid membrane. Taken from Pietzsch, J, 2004. The membranes of

eukaryotic cells contain, among others, three classes of lipids: glycerophospholipids,

sphingolipids, and cholesterol (CHO) or a closely-related sterol. Although the relative proportions

of these three lipid classes vary according to species or cell type, in vertebrates cholesterol is

typically present at levels of 30–40%, sphingolipids at levels of 10–20%, and glycerophospholipids

at levels of 40–60% of the total plasma membrane lipids (McMullen et al. 2004).

Page 12: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

12

The glycerophospholipids consist of a glycerol backbone, two ester-linked fatty acyl

chains, and a phosphorylated alcohol, typically phosphorylcholine (PC),

phosphorylethanolamine (PE) or phosphorylserine (PS). The fatty acyl chains usually

contain typically between 16 and 20 carbon atoms in total, being the number of

unsaturations the main difference between the different fatty acyl chains.

The sphingolipids are based on a more complex alcohol, the sphingosine. The

sphingolipids contains a single amide-linked fatty acyl chain, which is usually saturated

and may contain up to 24 carbon atoms, and either a phosphorylated alcohol (usually

phosphorylcholine), or one or more sugar molecules linked to the hydroxyl terminus of

the sphingosine backbone.

Finally, cholesterol consists of a fused cyclic four-ring structure containing a single polar

hydroxyl group and an isooctyl side chain, the cyclic ring system being essentially planar

and rigid.

In reference to the heterogeneity of the membrane, the asymmetry in the lipid

composition of the membrane is well demonstrated: the inner mono-layer contains a

higher concentration of PS and PE, whereas the extracellular mono-layer is enriched in

PC and sphingomyelin (SM) (Meer et al. 2011). More complex structures as lipid rafts,

caveolaes or clusters offer an additional level of complexity.

Depending on the lipid composition and distribution, the lipids are organized in different

phases or structures that present characteristic biophysical properties, such as fluidity,

electric charge, cross sectional area, lateral pressure profile, surface packing and non-

lamellar-phase propensities.

Page 13: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

13

Figure 3 . Membrane lipid structures. Examples of the relationship between lipid shapes, intrinsic

curvatures and lipid phases. A. Lipids with rectangular shapes (e.g. PC, SM) do not confer a

curvature strain forming lamellar phases. B. Lipids with a bulky polar head and only one acyl chain

(e. g. lisophospholipids) have an inverted cone shape inducing a positive curvature strain in

membranes. C. Lipids with a small polar head (e.g. PE, CHO, DAG) have a molecular shape that

resembles a truncated cone. They induce a negative curvature strain. D. Examples of phospholipid-

induced curvature strains in the membrane bilayer. From Lladó et al. 2014.

The most common lipid organization in the membrane is the lamellar phase which can

be subdivided in several types of lamellar sub-structures. These sub-structures can

change from one to another depending of lipid composition, pH, ionic strength, water

concentration or lateral pressure and the temperature that modulates the fluidity (Cullis

et al. 1979)

The lamellar α sub-structure (Lα), also known as fluid lamellar phase, liquid crystalline or

liquid disordered (Ld), is the structure found in most domains and regions of the cell

membrane, it is characterized by high quantities of PE and similar lipids, resulting in a

weaker surface pressure because the small polar heads of these lipids produce a lower

surface packing density and greater lipid and protein mobility. This lamellar structure can

evolve to a variety of more organized and less mobile structures, such as the gel phase

Page 14: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

14

(Lβ), pseudo- crystalline (Lc), ripped (Pβ) membranes and the solid ordered (so or Lo)

(Yeagle, 2005).

In the LB state, the phospholipid hydrocarbon chains are in the fully extended all-trans

conformation, the thickness of the phospholipid bilayer is maximal, and both intra- and

intermolecular motion are more restricted than in La phase.

The Lo phase is only possible in the presence of CHO. CHO acts increasing the mechanical

rigidity and cohesiveness and reducing the permeability of phospholipid bilayers, also

reducing the rotational and lateral diffusion rates (Lladó et al. 2014). CHO is also a key

component of the lipid rafts. Lipid rafts are enriched in CHO and SM with the acyl chains

of the lipids extended and highly packed. The lipid structure can operate as regulator for

protein interactions, and for these reason, essential cellular regulation processes are

compartmentalized in these CHO and SM raft domains and not out of them (Lingwood

et al. 2010; Pike et al. 2006).

In addition to the lipids that form fluid and organized lamellar phases, the cell membrane

may form non-lamellar phases, induced by non-lamellar-prone lipids such as PE, CHO,

diacyl-glycerol (DAG) and acidic PS. These lipids are structurally characterized by a non-

cylinder shape, usually exhibing truncated or inverted cone shapes, which induces a

curvature stress into one of the layers of the cell membrane (Figure 3). If non-lamellar-

prone lipids are abundant, the membrane can adopt a conformation in which some lipids

adopt an extended shape with one of the acyl chain out of the bilayer allowing a better

access to the inner part of the cell membrane (Ibarguren et al. 2013). The curvature stress

induced by the non-lamellar-prone lipids is usually organized into hexagonal and cubic

phases among others (Luzzati et al. 2009).

As this long introduction has tried to reflex, the membrane is a complex structure, key as

an element for the cell concept, but also allowing the communication between the

surrounding and the cell, including the communication between cells.

1.3 Autophagy

As happens with proliferation, every process in the cells is wonderfully under control. The

controlled degradation of cytoplasmatic material, as macromolecules or even organelles,

is known as autophagy. There are three main types of autophagy, depending on the way

in which cytoplasmatic material are delivered to lysosomes: chaperone-mediated

autophagy, microautophagy and macroautophagy.

Page 15: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

15

Autophagy plays an essential role in normal and pathological conditions such as

starvation, clearance of intracellular proteins and organelles, development, anti-aging,

elimination of microbes, cell death and tumor suppression. For this reason, autophagy is

now a marked target in cancer studies. Moreover, autophagy defects have been

associated with a wide range of disease, from microbial infection to chronic liver disease,

obesity, inflammatory bowel disease (IBD), aging, metabolic syndromes, Crohn’s disease,

Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and cancer (Rabinowitz &

White, 2010 ; Chen & Karantza 2011 ; Chen & White, 2011; Cheung & Ip, 2011).

The autophagy process (Figure 4) starts with a portion of the cytoplasm containing

material being engulfed by an isolation membrane called phagophore, which complete

elongation results in the formation of double membrane structure known as

autophagosome. These autophagosomes, containing the cytoplasmic material to be

degraded, are fused with lysosomes forming autolysosomes, which enzymes will degrade

the cytoplasmic material of the autophagosomes. Finally, the breakdown products are

released into the cytosol by permeases to recycle to use them in the cellular metabolism

(Rubinsztein et al. 2011)

Figure 4. Schematic diagram of the steps of autophagy. Autophagy begins with the formation of the

phagophore or isolation membrane. Phagophore elongation forms an autophagosome. The autophagosome

can engulf bulk cytoplasm. When the outer membrane of the autophagosome fuses a lysosome, it forms an

autophagolysosome. Finally, the sequestered material is degraded inside the autophagolyosome and

recycled. Meléndez & Levine, B, 2009

There are two major regulating pathways which control autophagy: The main regulating

mTOR dependent and mTOR independent.

The mTOR is the mammalian ortholog of the yeast protein kinase target of rapamycin

(TOR), which negatively regulates autophagy. In normal or non-pathological conditions,

when nutrients are present, the organism produce insulin and growth factors as signal of

nutrient abundance. The mTOR receive signals from insulin or growth factors from the

Page 16: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

16

class 1 phosphatidylinositol-3-OH kinase (PI3K-I). PI3K-I, using the plasma membrane

lipid phosphatidylinositol-4,5-bisphosphate (PIP2) produce phosphatidylinositol-3,4,5-

trisphosphate (PIP3). The PIP3 recruits phosphoinositide-dependent kinase1 (PDK1),

phosphoinositide-dependent kinase 2 (PDK2) and protein kinase B (AKT/PKB) from the

cytosol to the plasma membrane (Mizushima & Levine, 2010). PDK1 and PDK2 function

is the phosphorylation of the third serine/threonine kinase, AKT. The phosphorylation in

specific amino acids (Thr 308 phosphorylated by PDK1 and Ser 473 phosphorylated by

PDK2) results in the activation of AKT. Activated AKT finally inactivates by

phosphorylation the tuberous sclerosis complex (TSC) 1/2. The inactivation of TSC 1/2

leads to activation of Rheb protein which subsequently activates mTORC1. The activation

of mTORC1 leads to inhibition of autophagy (Mizushima & Levine, 2010)

Nutrient starvation or presence of rapamycin, results in mTORC1 inactivation (complex

of mTOR). The inhibition of mTOR results in the translocation from the cytosol to the

endoplasmic reticulum (ER) of the ATG1 complex, comprising Unc-51-like kinase 1/2

(ULK1/2), ATG13, focal adhesion kinase family interacting protein of 200 kD (FIP200) and

ATG101. This translocation leads to the recruitment of class III PI3K complex, consisting

of VPS34 (Vascular protein sorting 34), VPS15, Beclin -1 and ATG14 to the ER site (Easton

& Houghton, 2006). The formation of phagophores is initiated when FIP200 and ATG13

are phosporylated by ULK1 (Rabinowitz & White, 2010; Levine et al. 2011). For nucleation

phase, VPS34 is activated by Beclin-1 (Funderburk et al. 2010) to generate

phosphatidylinositol 3-phosphate (PI3P), activating two ubiquitin-like pathways for the

third phase, the elongation.

Figure 5. Autophagy is regulated by a set of autophagy-related proteins (ATG proteins). In the

absence of amino acids or in response to other stimuli, ATG1 and a complex of the class III PI3K

(phosphoinositide 3-kinase) VPS34 and beclin 1 lead to the activation of downstream ATG factors

that are involved in the initiation (a), elongation (b) and maturation (c) of autophagy. a In amino-

acid-rich conditions, VPS34 contributes to mTOR activation and inhibition of ATG1 and

Page 17: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

17

autophagy. b | The elongation and shape of the autophagosome are controlled by two protein

(and lipid) conjugation systems, similar to the ubiquitylation systems: the ATG12 and LC3

conjugation pathways, which include E1-activating and E2-conjugating enzymes. c | LC3

associated with the lumenal membrane remains trapped in the autophagosome and is degraded

during maturation into the autolysosome, which involves fusion of autophagosomes with late

endosomes, including endosomal multivesicular bodies and lysosomal organelles, and dissolution

of the internal membrane. VPS34 has a role in the formation of late endosomal multivesicular

bodies and lysosomal organelles contributing to the maturation stages of autophagy. Adapted

from: Levine & Deretic, 2007.

Atg 7, an ubiquitin-activating (E1-like) enzyme, activates the ubiquitin-like protein Atg12

through an ubiquitination-like process. Atg12 is subsequently transferred to Atg10, an

E2-like enzyme, which in turn conjugates with the lysine on Atg5 to create the conjugate

Atg5/Atg12. The conjugate complex binds to Atg16. The complex Atg12/5/16L is bound

to the outer layer of the isolated membrane of the phagophore (Levine et al. 2011),

enabling the second ubiquitin-like pathway to occur. The cysteine C-terminal residue of

the Atg8 (LC3) is cleaved by Atg4, a cysteine protease, to produce LC3I with a C-terminal

glycine residue. The cleaved LC3 I is conjugated to phosphatidylethanolamine (PE) by

Atg7 and Atg3 enzymes (Rabinowitz & White, 2010). This lipidated form of LC3 II is

attached to both outer and inner faces of phagophore. The cytoplasmic components are

recognized by p62/SQSTM1 and neighbor of BRCA1 gene 1 (NBR1) cargo receptor

proteins through the ubiquitin-interacting domains (UBA), and engulfed into the

phagophore by the interacting regions of LC3 (Gottlieb & Carreira, 2010). The

phagophore elongates to form an enclosed structure with double membrane known as

autophagosome, where target cargoes are engulfed in. At this moment, Atg12/5/16L is

released and Atg4 cleaves LC3 II from the outer surface of autophagosomes. The

autophagosome fuses with lysosome to form an acidified compartment by the help of

vacuolar proton ATPase (VPATPase). Finally, the cytoplasmatic materials of the

autophagosomes and the p62 and NBR1 proteins are degraded along with the other

components sequestered inside the autolysosomes of normal cells.

Notwithstanding the mTOR dependent is the main regulating pathway of autophagy,

there are also mTOR independent regulating pathways, being the phosphoinositol (PI)

signaling pathway the most important. In the PI pathway, the autophagy is negatively

regulated by intracellular level of free inositol and inositol 1,4,5-triphosphate (IP3) (Isakoff

et al. 2005) . The PI pathway is activated by G- protein coupled receptor after the

activation of the enzyme phospholipase C (PLC). PLC hydrolyzes PIP2 to form IP3 and

DAG. These IP3 are degraded by two enzymes, 5’-phosphatase and inositol

polyphosphate 1-phosphatase (IPPase), to form inositol monophosphate (IP1), finally

hydrolyzed by inositol monophosphatase (IMPase) into free inositol.

Page 18: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

18

1.4 Lipid membrane alterations in cancer

Similar to regulation of gene expression, changes in the presence and levels of

membrane lipids species have been described in several human pathologies, associated

either with adaptive responses or with the etiology of the disease. In this regard,

numerous studies have shown that the lipid composition of tumor cell membranes is

altered with respect to non-tumor cells. This area of study has received little attention in

cancer research, mainly because structural and functional concepts of lipid alterations in

cancer are more difficult to understand than the functional role of certain proteins and

their genes in defining cancer cell phenotypes. Although it has not been shown a

common pattern of alterations characteristic for different kinds of tumors yet, certain

cancer induced lipid prolife changes have been described and should possess some

diagnostic values (Michalak, 2003).

In this context, a hallmark of cancer cells is the constitutive activation of the fatty acid

biosynthetic pathway, which produces saturated fatty acids (SFA) and monounsaturated

fatty acids (MUFA) to sustain the increasing demand of new membrane phospholipids

with appropriate acyl composition (Kuhajda 2006, Rashid et al. 1997, Swinnen et al. 2000).

In this regard, the increased levels of oleic acid, detected in several tumors, are related

to the activation of the fatty acids synthesis (Igal, 2010). The enzyme responsible for the

oleic acid synthesis is the steaoryl-COA desaturase (SCD).

Gangliosides, which are membrane-bound glycosphingolipid molecules, are frequently

aberrantly expressed in tumors (Hettmer et al. 2005). Ganglioside antigens on the cell

surface act as immunosupressors, and certains gangliosides, such as GD3 or GM2,

promote tumor associated angiogenesis (Birkle et al. 2003). The reduced levels of

Ceramide that were found in some types of cancer could be related to its pro-apoptotic

role (Riboni et al. 2002). One lipid alteration highly connected to the genetic alterations

found in cancer, is the elevated levels of PI(3,4,5)P3, formed by the activation of PI3K that

was observed in several tumors and contribute to oncogenesis through the PI3K/AKT

pathway (Vivanco & Sawyers, 2002)

It has been demonstrated in several studies that different types of lipids and their relative

abundance in the cell membrane can control numerous functions and regulate the

activity and localization of membrane proteins (Escribà et al. 1996;, Escribà et al. 1997;

Vögler et al. 2004). In the case of cancer, it has been shown that the proportion of

membrane lipids is altered in cases of breast, lung, pancreas, liver, prostate, brain and

colon cancer (Mikirova et al. 2004; Michalak et al. 2003).

Page 19: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

19

1.5 Membrane-Lipid Therapy

As indicated, several studies have related a high number of important diseases with

structural or molecular disarranges at the lipid membrane, from where the most of the

pathways that regulate cellular functions began.

The role of proteins in the development of diseases is well known. Perhaps the most

direct relationship is in monogenic diseases where a single altered gene produces an

altered protein, with non-function or over-function which produces the disease. In the

case of multifactorial diseases, although there are more factors, it is known that part of

disease is due to the performance or non-performance of different proteins.

From the point of view of classical molecular medicine, the focus has always been to

develop drugs that target a protein that has been determined to be key to the disease

process in particular. This strategy has proven to be effective in many diseases and a very

high percentage of treatments are included in it. However, it has also shown that in

complex diseases, such as cancer, at the molecular level the mismatches are so abundant

that affecting one or two proteins with a drug in a try to reverse the disease,

unfortunately, is no sufficient.

Figure 6. Main difference of the two biological approaches mentioned in this master thesis for

the treatment of human pathologies. The molecular entities regulated by the treatment are

colored, whereas the molecular entities that are not affected by the therapy are shown as open

symbols. (a) Conventional chemotherapy is characterized by the interaction of a drug with a target

protein (gray). Upon drug binding, the activity of such a protein, the downstream elements and

gene expression are modulated (b) In membrane-lipid therapy, the clinical drug binds to

membrane lipids, regulating the structure of the membrane, with subsequent modulation of the

activity of a membrane protein and downstream events. Adapted from: Escribá, 2006).

If we consider that most of the cancer-related pathways are upstream activated in the

membrane and the lipid modifications occurring in cancer cells are associated with the

activation of proliferation and tumorigenesis, it is conceivable that lipid modifications

can regulate these pathological cell signaling pathways. The lipids control the interaction

Page 20: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

20

and activity of many proteins, and not only that, the proteins bonded to the membrane

may alter the structure of this, being reciprocal the regulatory effect between proteins

and lipids.

Given the importance of the membrane, the strategy to develop specific therapies to

regulate the lipid membrane structure for the treatment of several diseases arising. The

Membrane-Lipid Therapy (MLT) seeks to regulate the participation of membrane lipids

in cellular functions by using lipid product being intercalated in the membrane and

regulate its structure, and therefore modifying the location and activity of membrane-

interacting proteins (Figure 6) (Reviewed in Escribá, 2015).

The relation between lipid structure and function, clearly known and accepted in the

protein world but not in the lipidic one, is the starting point for the rational development

of synthetic lipid compounds as effective therapeutic drugs. In addition, the lipid drugs

developed and tested on this group show a low toxicity profile, being the collateral

effects or toxicity of several cancer treatments a main trouble to overcome.

To demonstrate the efficacy of this novel approach, we can take as an example the

Minerval, a fatty acid analog to the oleic acid designed by Dr. Escribá, which works by

activating the sphingomyelin synthase (SMS). Currently Minerval is in Phase I / II clinical

trials (clinicaltrial.gov identifier NCT01792310) for the treatment of glioma and other

solid tumors. Preliminary data in those human studies are promising; several patients in

which the standard treatment had not worked showed a partial response or stable

disease after Minerval administration. As also mentioned, the toxicity profile of Minerval

is really positive, not having seen any SAE (Serious Adverse Event) at even high dosed

(up to 12 grams per day).

This master thesis can be included in the Membrane-Lipid Therapy, because the object

of study have been several synthetic triglycerides.

1.6 Objects of study: Mimetic triglycerides (TGMs)

Triglycerides are structures formed by a glycerol backbone and three fatty acid chains

attached via an ester bond (Figure 7). Physiologically involved in glucose metabolism and

fat, their presence at high levels is commonly used in clinical practice as marker of

atherosclerosis or heart disease risk.

When triglycerides are ingested with the diet, due to their large size, they cannot be

absorbed in the duodenum or enter cells until pancreatic lipase breaks the ester bond,

releasing the fatty acid chains. Thus, the absorbed product can be either free fatty acids,

monoglycerides (a glycerol molecule attached to a fatty acid) or diglycerides (one

Page 21: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

21

molecule of glycerol with two fatty acids) which enter the cell through the FAT receptor.

Once in enterocytes, the triglycerides are re-formed from their fragments, and bind to

CHO and other proteins to form part of chylomicrons.

Figure 7. Simple representation of the structure of a triglyceride. A triglyceride is formed by a

glycerol backbone where three fatty acids chains are bonded. For this master thesis rationally

designed triglycerides has been used. The modifications where performed on the fatty acids

chains, even developing molecules with three different modified fatty acids chains.

The antitumor role of triglycerides is understood through the known antitumor effect of

some fatty acids that may be part of triglycerides. More than 10 years ago the effect of

fatty acids lauric, stearic, palmitic, oleic, linoleic, alpha-linolenic, gamma-linolenic,

arachidonic, docosahexaenoic and eicosapentaenoic was studied in different lines of

pancreatic cancer (Falconer et al. 1994). All the polyunsaturated fatty acids (PUFA) tested

had an inhibitory effect, with EPA being the most potent. SFA and MUFA fatty acids were

not inhibitory. Another clear example that shows the relation between structure and

function.

Triglycerides become very interesting objects of study when their potential use as

modified fatty acids carriers, such as Minerval, is exploited. The fact that a triglyceride

can carry up to three identical modified fatty acids with remarkable antitumor effect is

interesting, like happens in simple TGMs. The complexity and interest of their study

increases when we add two or three species of different fatty acids to the same molecule

of glycerol, all with a particularly physiological and therapeutic effect, and that is the case

of mixed TGMs.

Thus, this group has developed a battery of modified fatty acids that enhance their

antitumor activity. Furthermore, we have developed these triglycerides containing fatty

acids either separately or in mixed form, each possible combination results in a different

molecule with specific therapeutic effect against several pathologies, like cancer. The

antitumor efficacy against pancreatic cancer of these mimetic triglycerides or TGMs has

been the object of this master thesis study.

Page 22: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

22

2. Objectives

The general aim of this master thesis was the investigation of the possible antitumor

activity of different lipid compounds developed by the Molecular and Cellular

Biomedicine group. Most of these compounds were TGMs or mimetic triglycerides,

triglycerides the fatty acids chains of which were modify under rational design.

Several compounds were screened for their antitumor potential in pancreatic

adenocarcinoma cell line Mia-PaCa-2. Of the analyzed compounds, the most interesting,

TGM4 and TGM5, were investigated in vivo with Mia-PaCa-2 xenograft tumors on

immunosuppressed mice. Finally, the mechanism of action of the TGM4 was studied.

The particular objectives of this master thesis were:

1. Determine the potential antitumor activity of the TGMs compounds developed by the

research group where this work was conducted.

2. Study the toxicity after chronic treatment with the compounds TGM4 and TGM5.

3. Determine the antitumor capacity of the compounds TGM5 and TGM4 in vivo, using a

human pancreatic tumor xenograft model.

4. Explore how the TGM4 exerts its antiproliferative effect; for this, three approaches were

planned:

4.1 Determine of the cell cycle alterations produced by TGM4 in pancreatic tumor

cells.

4.2 Investigate the TGM4 effects in pancreatic tumor cells through different key

proteins for the survival or cell proliferation, determining the mechanism through

TGM4 causes cell death.

4.3 Study of the lipid alterations produced by TGM4 in pancreatic cancer cells.

Page 23: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

23

3. Materials and methods

3.1 Cell lines

Mia-PaCa-2 (Human pancreatic carcinoma) and PANC-1 (human pancreatic carcinoma

of ductal cells) were bought from ATCC (American Type Culture Collection).

3.2 Cell culture

3.2.1 Cell lines and growth conditions

Mia-PaCa-2 and PANC-1 monolayer cell lines were maintained and grown in 75 cm²

flasks with DMEM (Dulbecco's Modified Eagle Medium) with phenol red including 10%

of fetal bovine serum (FBS), 100 U/ml of penicillin and 100 µg/ml of streptomycin. The

medium was also supplemented with D-glucose (4.5 g/L), L-Glutamine (4 mM) and

Sodium pyruvate (1 mM).

They were incubated in HEPA filtered cell incubator (Memmert GmbH Co, UK) at 37°C

with 95% humidified air and 5% CO2. Cell culture experiments were carried on laminar

vertical flow cabinet (Telstar S. A., Terrasa, Spain).

3.2.2 Thawing

Frozen cells in cryovials taken from nitrogen tank in cold room were defrosted at room

temperature. Then, they (2 ml) were transferred into T25/T75 tissue culture flask. The

volume was completed to 5/15 ml with DMEM complete medium and flasks were

incubated in CO2 incubator.

3.2.3 Cell passaging / maintaining

When the cells in flask reached 80% of confluence, the medium was discarded. The T75

flask was washed with 5 ml of PBS (137 mM NaCl, 2.7 mM KCl, 12 mM Na2HPO4 and 1.38

mM KH2PO4) in order to remove waste materials and serum which includes trypsin

inhibitors. Then 2 ml EDTA-trypsin was added and the flask was incubated in CO2

incubator for 2-5 min. When detachment was observed under inverted light microscopy,

flask was taken from incubator and growth medium was put in immediately to stop

trypsin activity, which large time exposure can cause damage in cells. Then the cells were

divided in the desired number of flasks, normally in 1:3-1:6 dilutions.

Page 24: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

24

3.2.4. Freezing

If freezing of the cells was required, after trypsinization of cells, they were centrifuged at

600 x g for 5 minutes. The supernatant, which includes medium, dead cells and waste

products, was discarded. Pellet includes cells was re-suspended with freezing medium.

Freezing medium was prepared by mixing 10% of dimethylsulfoxide (DMSO), a

cryoprotectant that lowers the freezing point, and 90% FBS.

Cell suspension was put in cryovials, 2 ml of suspension for each. The cryovials were

stored at – 80°C to achieve gradual freezing and after three days they were placed in

liquid nitrogen tank (-190°C) for long term storage.

3.2.5 Cellular treatments

To dissolve the TGMs (lipid compounds), a stock solution of 100 mM of TGM was

prepared in full DMSO (Polar aprotic solvent). Then the stock solution was dissolved in

full DMEM medium to get the desired concentration, never surpassing the 0.5% of final

DMSO in the medium.

3.3 Cell proliferation / cytotoxicity studies

For cytotoxicity experiments, Mia-PaCa-2 and PANC-1 cell lines were seeded at a density

of 3x103 cells/well into 96 well plates and incubated at 37°C for 24 hours in CO2 incubator.

After 24h, when cells were attached, the medium was changed and replaced with new

medium containing one of the different TGMs studied. Different molecules were studied

at different times, the following table (Table 1) resume them.

TGM0 TGM5 TGM25

TGM1 TGM6 TGM46

TGM2 TGM12 TGM146

TGM4 TGM16

Table 1. List of the different molecules used for the studies of this master thesis, all forming part

of the TGMs library.

The cytotoxic effects were studied with Cell Proliferation XTT Kit (Roche Diagnostics, S.L.

Applied Science, Barcelona, Spain). The basis of this technique is the reduction of

tetrazolium salt XTT by living metabolically active cells to an orange colored formazan, a

reaction produced by succinate dehydrogenase enzymes of mitochondria respiratory

chain (Figure 8). Only the viable cells with intact mitochondrial and cellular membrane

Page 25: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

25

have active dehydrogenases, thus the concentration of formazan formed is proportional

to the number of living cells. These bio-reduction of the tetrazolium salt is related with

the production of NAD(P)H through glycolysis.

Figure 8 Cell Proliferation Kit XTT employs 2,3-Bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-

tetrazolium-5-carboxanilide salt (XTT). Only in living cells mitochondria are capable to reduce XTT

to form an orange colored water soluble dye. Therefore, the concentration of the dye is

proportional to the number of metabolically active cells. Source: AppliChem

After incubate the cells the desired time with the different TGMs, the medium was

replaced with DMED without phenol red (it interferes on the absorbance of the formazan

product) mixed with the reagents XTT and electron-coupling, following manufacturer‘s

instructions. Then the cells were incubated at 37ºC until the color compound was formed.

Finally, the absorbance was read at a wave length of 495 nm in a plate reader (FLUOStar

OMEGA, BMG LABTECH, Germany)

For statistical confidence, 2 or 3 independent experiments were performed for each TGM

and each one was quadruplicate wells. According to dose-response curve (log (inhibitor)

vs. normalized response – Variable slope model) drawn according to percent viability, the

dose necessary to kill the 50% of the cells (IC50, inhibitory concentration 50) was

calculated with Graphpad Prism Version 5.

3.4 Cell cycle studies

The analysis of the effect of the TGM4 was performed through flow cytometry. PANC-1

cell lines were seeded at a density of 250.000 cells into 6 cm of diameter plates and

incubated at 37°C for 24 hours in CO2 incubator. After 24h the medium was changed and

replaced with new medium containing TGM4 at the desired concentration (20, 30, or 40

µM). The cells were incubated during 6, 12, 24, or 48 hours.

Page 26: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

26

After the desired time of incubation, the cells contained

in the medium, both alive and dead were recovered by

centrifugation at 1000 x g for 5 minutes in 5 ml cytometry

tubes and resuspended in complete media. Then, the live

and attached cells were detached with EDTA-trypsin,

combined with the floating ones and centrifuged 1000 x

g for 5 minutes. The supernatant was totally discarded

and all the cells were stored at the tube.

To fix the cells, cold 70% ethanol was slowly added to the

tube while vortexing. To avoid the clumping of the cells

the solution was thoroughly pipetting. The tube was let

O/N at 4ºC. 24h later, the ethanol was discarded after a centrifugation at 2500 x g and

4ºC for 5 minutes. At this point PBS can be added to store the cells at 4ºC for at least a

week. To perform the cytometry the cells were washed with 1 ml of sodium citrate 38

mM pH 7.4. Then the sodium citrate was discarded after centrifuging at 2500 x g and 4ºC

for 5 minutes.

Finally, the cells were re-suspended in 500 µl of “Buffer A” solution formed by sodium

citrate 38 mM pH 7.4, 50 µg/ml of propidium iodide and 5 µg/ml of RNase A (Sigma-

Aldrich Co, St Louis, MO) and incubated at 37ºC for 20 minutes. After the addition of

propidium iodide the tubes were maintained permanently in the darkness until the

cytometry lecture. The flow cytometry was performed in a flow cytometer Beckman

Coulter Epics XL (Beckman Coulter S.A, Madrid, Spain). The different cell populations

corresponding to the different phases of the cell cycle (Sub-G1, G0/G1, S and G2/M) were

defined through their DNA quantity (Figure 9). The results were analyzed with the

software FlowJo (FlowJo, USA).

3.5 Western Blot studies

3.5.1 Cell lysate

For the immunoblot studies, 325.000 PANC-1 cells were seeded into 6 cm diameter plates

and incubated at 37°C for 24 hours in CO2 incubator. After 24h the medium was changed

and replaced with new medium containing TGM4 at a concentration of 30 µM.

At the desired time of treatment, the medium was discarded and the plates were washed

twice with cold PBS 1X and frozen at -80ºC until all the plates were collected. Then, 300

µl of lysis buffer (20 mM HEPES, 2 mM EDTA, 0.5 mM EGTA, 1.5 mM MgCl2, 1 mM

Figure 9. Relationship

between the cell cycle and the

DNA histogram. Source:

Ormerod & Novo, 2008.

Page 27: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

27

cantaridine, 1 mM ortovanadate and a protein inhibitor cocktail from Roche) were added

to the plate. After 5 minutes from the addition of the lysis buffer, the cells were scrapped

and the lysate containing the protein was transferred to a new tube. To fully homogenize,

the samples were sonicated on ice twice for 5 seconds.

3.5.2 Protein quantification

The protein concentration determination of each sample was performed with the protein

quantification DCTM kit (Bio-Rad, Barcelona, Spain), a colorimetric assay used for the

quantification of proteins in presence of reducer agents and detergents. The DCTM

method is a modification of the classical Lowry method, based on the reduction of the

reagent Folin phenol, that modifies the color of the reagent, and changes are quantified

measuring the absorbance at 750 nm (Lowry et al. 1951)

The DCTM kit contains 3 reagents: A, S and B. The quantification was performed in a 96-

well plate. For each sample, triplicates were done. 5 µl of the sample were added to the

well, then 25 µl of A' reagent (mixing reagents A and S in a proportion 50:1) were added

to each well. Finally, 200 µl of the reagent B were added to the wells and then the plate

was maintained at room temperature for 15 minutes. Finally the plate absorbance was

read in a plate reader at 750 nm.

Protein concentrations were obtained by interpolation of the absorbance values on the

standard curve made with known concentrations of bovine serum albumin (BSA), from

0.2 to 6 mg/ml. Once the protein concentration of the samples was known, samples of

equal protein concentration were prepared to perform the experiments, diluting the

necessary samples with protein lysis buffer until achieve the desired concentration.

Protein samples were stored in ice or frozen at -20ºC to avoid the degradation during

the whole process.

3.5.3 Western Blot

Samples were mixed with loading buffer (Tris-HCl 12 mM pH 6.8, β-mercaptoethanol [-

ME], 1%, SDS 0.2%, bromophenol blue 0.01%, and glycerol 50%) in a 1:10 proportion and

boiled at 95ºC for 5 minutes. Then, 30 µg of whole cell lysate of each sample were loaded

on a 10 % SDS polyacrylamide gel. As a protein ladder standard, 2.5-5 µl of Precision Plus

Protein all blue standard (Bio-Rad) was loaded.

The acrylamide concentration in the gel determines the range of separation of proteins

during electrophoresis. These gels have two distinct areas, an area for concentration gel,

where the samples are loaded, and a zone for separation gel at the bottom, where the

Page 28: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

28

samples are separated. The gel concentration ("stacking") is composed of acrylamide-

bisacrylamide 4%, Tris-HCl pH 6.8 166mM, SDS 0.1%, ammonium persulfate 1% and N,

N, N ', N'-tetramethylethylenediamine 0.1%. Gel separation ("resolving or running") is

composed of acrylamide-bisacrylamide 9.5%, Tris Base pH 8.8 1M, SDS 0.1%, ammonium

persulfate 0.4% and N, N, N ', N'- tetramethylethylenediamine 0.04%.

After loading all the samples in the SDS-polyacrilamide gels, SDS-PAGE (sodium dodecyl

sulfate-polyacrilamide gel, electrophoresis) was performed. (Laemmli, 1970).

The SDS, together with the -ME, has the ability to denature the proteins. The SDS gives

net negative charge to the protein allowing them to migrate through the gel

proportionally to its mass, since there is a constant charge/mass ratio (approximately 1

molecule of SDS per 2 amino acids, with a ratio SDS/protein of 1.4 g/g). . On the other

hand, the -ME breaks the disulfide bonds in the protein separating its subunits.

The electrophoresis was set at 90 V for the stacking phase and changed to 120-140 V for

the running phase. The electrophoresis buffer is composed of 19.2 mM Tris-base, 0.19 M

glycine pH 8.6, and 0.1% SDS. When the electrophoresis was finished, the proteins into

the gel were transferred to a nitrocellulose membrane (GE Healthcare, Kent, UK). The

transfer process was performed in cold conditions and applying a constant amperage of

350 -400 mA for 2 hours with buffer consisting of 19.4 mM Tris-base and 0.19 M glycine

and 20% ethanol.

After finishing the transfer, the membrane was blocked in 5% skim milk in TBS (50 mM

Tris-Cl, pH 7.6; 150 mM NaCl) for 30 minutes in order to prevent non-specific antibody

binding. Once blocked, the membrane was incubated in a primary antibody dilution

(1:1000) against the protein of interest and allowed to stir overnight at 4ºC. Primary

antibody solution was prepared containing 5% BSA and 0.1% Tween 20 (Sigma-Aldrich

Co., St. Louis, MO). The next day, the solution was removed of the membrane and 3

washes were performed for 5 minutes with TBS and Tween 20 0.1%. The membrane was

incubated for 1 hour in the dark with the secondary antibody (dilution 1: 5000)

conjugated with a fluorochrome (IRDye 800CW Donkey Anti-Mouse IgG (H + L) or IRDye

800CW Donkey anti Rabbit IgG (H + L), LI-COR Biosciences, USA). The secondary

antibody was prepared in 2.5% skim milk in TBS with 0.1% Tween 20. Then, the secondary

antibody was removed and the membrane was washed twice for 5 minutes TBS and 0.1%

Tween 20 and once with TBS for 5 minutes. Since the incubation with secondary antibody

the process was carried out maintaining the membrane in darkness.

The membranes were scanned in near infrared spectroscopy (Odyssey Infrared Imaging

System, LI-COR, Inc., Lincoln, NE, USA) with a resolution of 84 microns and analyzed with

Image StudioTM software (LI-COR, Inc., Lincoln, NE, USA) obtaining the values of

Page 29: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

29

integrated optical density (DOI) of each band. The α-tubulin content in each sample was

used as a loading control.

3.6 Thin Layer Chromatography

The analysis of the TGM4 effect on cellular lipid composition was studied through Thin-

Layer Chromatography (TLC). TLC is based on the separation of a mixture of compounds

as they migrate with the help of a suitable solvent through a thin layer adsorbent material

which has been applied to an appropriate support. Different stationary phases and

several combination of polar/organic solvents are available to separate the mixture by

different characteristics. However, adsorption is the most common mechanism of

separation, where the sample is continually fractionated as migrates through the

adsorbent layer.

Competition for active adsorbent sites

between materials to be separated

and the developing solvent produces

continuous fractionation. A portion of

the material to be separated will be

found in the mobile phase and a

portion will be adsorbed to the solid

adsorbent particles. As the process

continues the several components move

different distances, depending on their

relative affinities for the adsorbent as compared with the migrating solvent (Figure 10).

The affinity of the components is mainly related to their polarity. The more polar

compounds are held back by the adsorbent while the less polar compounds advance

further. In these experiments silica (as adsorbent) pre-coated plates were used.

To the procedure, 300.000 PANC-1 cells were seeded into 6 cm diameter plates and

incubated at 37°C for 24 hours in CO2 incubator. After 24h the medium was changed and

replaced with new medium containing TGM4 at concentrations of 20 µM and 30 µM. For

the control, new medium without TGM4 was added. The cells were incubated during 12

and 48 hours, then the medium of the plated was discarded and the plates were washed

and frozen until their use.

3.6.1 Lipid extraction

For each plate 600 µl of hypotonic buffer (20 mM Tris pH 7.5 and 1 mM EDTA) was added

before cells were thawed. Then the plates were scrapped and the cells in the hypotonic

Figure 10. Scheme of a Thin Layer

Chromatography

Page 30: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

30

buffer cell extracts were collected in 1.5 ml tubes. To fully homogenize the samples, they

were sonicated on ice 3 times 10 seconds on/10 seconds off at 20% of amplitude.

Neutral lipids or generally storage lipids are extracted with relatively non-polar solvents

as chloroform and/or petroleum ether, but membrane-associated lipids (the most

interesting for the MLT) requires polar solvents such as methanol to disrupt hydrogen

bindings or electrostatic forces. A mixture of Chloroform: Methanol (2:1) was used to the

lipid extraction for these experiments. To avoid the peroxidation of the extracted lipids,

all solvents were peroxide-free HPLC grade.

Then, 550 µl of each sample were transferred to a new glass tube, and 2.75 ml of a

chloroform: methanol mixture (2:1) was added. The samples were mixed by vortexing

and centrifuged for 10 minutes at 500 x g and 4ºC. After the centrifugation, three parts

were clearly observed, from the top to the bottom, the aqueous phase, a thin film formed

by the proteins, and the organic phase where the lipids were. The organic phase was

collected carefully, to avoid drag other components that will affect the separation of the

lipids in 2 ml tubes. The tubes were placed in an argon evaporator to evaporate the

organic solvents and, after that, the tubes containing only the lipids were stored at -20ºC.

3.6.2 Thin layer chromatography

Whatman silica gel-60 plates (20x20 cm, 250 µM, GE Healthcare, England) were heat-

activated at 110ºC for 1 hour, then the lipid samples where re-suspended in 40 µl of

chloroform and streaked onto the plates, this process was repeated twice. Phospholipids

were separated using chloroform/methanol/acetic acid/water (60:50:1:4 by volume)

which separated all major glycerophospholipids. Lipids were identified using

commercially available standards (Larodan, Sweden). To develop, plates were air-dried,

submerged in a solution containing 5% (W/v) H3PO4 and 4% (W/v) CuSO4, and charred

at 180ºC for 10 minutes (Gellerman et al. 2005). Lipids were finally quantified by photo-

densitometry using to the Quantity one software (Bio-Rad).

3.7 In vivo studies

3.7.1 Experimental animals

The animal model used to conduct the experiments was the immunodepressed NUDE

mice (Swiss Crl:NU (Ico)-Foxn1 nu; Charles River laboratories, France), aged between 4-6

weeks and with an approximated 25 grams of weight.

Page 31: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

31

These animals, due to the lack of thymus and, consequently, their immunodepression

state, were maintained always under sterile conditions, kept on plastic cages located on

a sterile closet (EHRET, Labor_U_Pharmatechnik, Deutschland) with a constant

temperature of 28ºC. The cabinet was kept in a room with a 12 hour light/12 hour dark

schedule and a relative humidity of 40-60%. All the work or manipulation of the animals

was conducted in every moment under sterile conditions on BSL-2 flow cabinets.

3.7.2 Determination of the toxicity of the compounds TGM4 and TGM5.

To study the toxicity and to establish the higher but safer dose of each compound, a

toxicity study was designed and performed. Different and increasing doses of TGM4 and

TGM5 were administered to BALB/ NUDE heterozygous mice by oral cannulation. The

doses were: 50 mg/kg, 100 mg/kg, 250 mg/kg, 500 mg/kg and 1000 mg/kg. The

compounds were mixed with soy oil when necessary to get the desired concentration.

Each dose was tested on 3 animals for 15 days, during which the animals were treated

and weighed daily. The weight of the animals was used as the first indicator of toxicity.

The behavior of the animals was also observed as an indicator of toxicity.

The diet during the 15 days of treatment was a standard one based on food pellets and

water ad libitum. After the treatment, all the animals were euthanized by decapitation

and dissected to collect different organs (brain, spleen, lung, heart, kidneys and liver) to

anatomical pathology or compound distribution studies.

3.7.3 Antitumor studies

3.7.3.1. Subcutaneous inoculation of tumor cells

To study the in vivo effect of the chosen compounds, a xenotransplant model was used.

The cell line Mia-PaCa-2 was expanded in 15 cm plates. Once achieved the desired

number of plates or cells, the plates were washed with PBS and the cells were trypsinized

and collected in DMEM medium. Then, the viable cells density of the suspension was

determined by counting them on a Bürker camera. Once the concentration of cells was

calculated, they were centrifuged 5 minutes at 600 x g to discard the complete DMEM

medium and they were re-suspended in DMEM medium without fetal bovine serum, to

avoid immune reactions even possible on immunodepressed animals.

A total of 7.5 x 106 cells were injected subcutaneously with a 25G caliber needle, in a

volume of 150 µl. 1 week after inoculation, the tumors had an approximated size of 5

mm and the treatments began.

Page 32: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

32

During the treatment the tumors size was measured once a week with a digital caliper.

We also measured once a week the weight of all the animals as an indicator of toxicity.

The tumor volumes were calculated by the formula: Volume (mm3) = (2W x L) / 2, where

W represents the width and L represents the length of the tumor (Barbacci et al. 2003).

3.7.3.2 Animals treatments

To investigate the in vivo efficacy of the chosen compounds, TGM4 and TGM5, the

animals were divided into 4 groups: treated with TGM4, TGM5 or Gemcitabine and

control animals. A total of 8 animals (4 males and 4 females) was used for each condition.

The treated animals received daily 1000 mg/kg of TGM4 or TGM5, both by cannulation.

The control animals received nothing or 100 mg/kg of Gemcitabine twice a week.

Gemcitabine is the standard of care for the treatment of pancreatic cancer and it was

administrated through intraperitoneal injection. The duration of the experiment was 43

days.

After the treatments, the animals were euthanized by decapitation. Immediately, the

animals were dissected and the blood, the tumor, and several organs (brain, spleen, lung,

heart, colon, kidneys and liver) were collected to possible future studies. The blood was

stored at 4ºC and then centrifuged at 4ºC and 1000 x g to collect the plasma fraction.

The tumor and the organs were divided into two parts, one was frozen in liquid nitrogen

and the other one was preserved in formalin 10%.

All the protocols and procedures were revised and approved by the Comité Institucional

de Investigación Animal (Comisión de Bioética de la Universitat de les Illes Balears)

3.8 Data analysis

All data shown in the graphs correspond to the mean values ± standard error of mean

(SEM) of at least 2 independent in vitro experiments (each duplicated) or cellular

experiments. For animal studies there is indicated in the graphs the number of animals

used (N).

The statistical analysis was performed by the average t-student, configured as unpaired,

two-tailed test, with a confidence intervals of 95%. For the statistical analysis of the

animal studies, the non-parametric Mann-Whitney test was used. All statistical analysis

were performed using GraphPad Prism 5.0 program.

The differences between experimental groups were considered statistically significant at

p <0.5. The different significances were represented as: *, p <0.05, ** p <0.01, *** p

<0.001.

Page 33: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

33

4. Results

4.1 Mimetic triglycerides (TGMS) inhibit cell viability and proliferation.

A screening of the antitumor activity of the different TGMs, included in the library of

compounds rationally designed from the structure of the triglycerides, was performed.

The effect of the TGMs on the viability and proliferation of pancreatic cancer cell line

Mia-PaCa-2 was tested through the XTT assay.

Page 34: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

34

Figure 11. Effect of different TGMs in cell viability and proliferation of tumor cells Mia-PaCa-2.

Cells were treated with several concentrations of the indicated compounds for 72 hours. To

determine the IC50 the XTT cell viability assay was used. (A) TGM0 (n=12). (B) TGM1 (n=12). (C)

TGM2 (n=12). (D) TGM4 (n=12). (E) TGM5 (n=12). (F) TGM6 (n=12). (G) TGM12 (n=12). (H) TGM16

(n=12). (I) TGM25 (n=12). (J) TGM46 (n=12). (K) TGM146 (N=12).

Page 35: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

35

Figure 12. Effect of TGM1 (A) and TGM5 (B) in cell viability and proliferation of tumor cells Mia-

PaCa-2. Cells were treated with several concentrations for 24, 48 and 72 hours. The effect is time-

dependent. (n=12)

Figure 13. Effect of TGM4 (A) and TGM5 (B) in cell viability and proliferation of tumor cells PANC-

1. Cells were treated with several concentrations 72 hours. To determine the IC50 the XTT cell

viability assay was used. (n= 12)

All the TGMs tested proved to have antitumor activity, but there were differences about

their potency (Figure 11). The potency was measured using the IC50, which reflects the

necessary concentration of a compound to have the 50% of the alive cells regarding to

a non-treated control due to inhibition of proliferation or induction of cell death. The

Table 2 resumes all the molecules tested and their correspondent IC50 with a 95%

confidence interval.

Page 36: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

36

Molecule 95% Confidence Intervals -

IC50 (µM) Molecule

95% Confidence Intervals -

IC50 (µM)

TGM0 72,75 – 95,64 TGM12 2,94 – 4,25

TGM1 69,84 – 85,65 TGM16 17,20 – 21,52

TGM2 10,57 – 13,48 TGM25 2,40 – 3,52

TGM4 18,59 – 21,86 TGM46 7,94 – 10,05

TGM5 2,60 – 3,16 TGM146 3,60 – 4,71

TGM6 5,43 – 7,50

Table 2. Table summarizing the 95% confidence intervals of the IC50 at 72 hours of all TGMs tested

in vitro with Mia-PaCa-2 cells.

Another factor to consider evaluating the antitumor activity of a compound is its

efficiency. The efficacy is reflected by the ability of the compound to kill all the cells

present in the culture (achieving the 0% of cell survival). With this objective, several TGMs

were tested at a high dose of 200 µM and most of them achieved it, excepting TGM0,

TGM1, TGM2 and TGM6 (Figure 11).

The antitumor effect was shown to be dose-dependent in all cases. From the data

obtained with two compounds at different times of treatment, it can be estimated that

as expected, the effect of TGMs is also dependent on the time, so the longer the

treatment, the greater the effect obtained (Figure 12). The compounds TGM4 and TGM5

were tested on the pancreatic cell line PANC-1 (Figure 13). On PANC-1 both compounds

maintained the antitumor activity.

4.2 Determination of a safe dose to perform in vivo studies with TGM4 and TGM5.

Before performing in vivo studies to test the antitumor activity of TGM4 and TGM5

compounds, a toxicity study was required, to establish the appropriate dose to which the

mice will be treated. Different and increasing doses of TGM4 and TGM5 were

administrated daily to BALB / NUDE heterozygous mice by oral cannulation. Each dose

Page 37: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

37

was tested on three animals of mixed sexes for 15 days, during which the animals were

weighed and observed for toxicity indicators daily.

Figure 14. TGM4 effect on weight of BALB/nude mice during 15 days of treatment at different

doses. n=3

Figure 15. TGM5 effect on weight of BALB/nude mice during 15 days of treatment at different

doses. n=3

Page 38: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

38

The results showed that the treatment with TGM4 or TGM5 did not affect the weight

(Figure 14 and Figure 15) or the behavior of any animal. Their weight varied during the

15 days without significant differences over the control. The behavior, mainly reflected in

the locomotive activity and variations in normal intake of the animals, was not affected

by the treatment with TGM4 or TGM5.

From the data obtained on these toxicity studies indicated that the highest dose tested

seems to be safe. Owing to that it was used for the antitumor in vivo studies and the

mice were treated at a dose of 1000 mg/kg.

4.3 TGM4 effect on the progression of Mia-PaCa-2 cell line xenograft in nude

immunodepressed mice

The cell studies are always necessary and the basis to test the antitumor activity of new

molecules, but the huge differences in complexity found between a cell culture and a

complex live animal as the rodents are, can make a big difference, therefore, the TGM4

and TGM5 antitumor activity was tested in vivo on mice. A model of human pancreatic

tumor xenograft in immunosuppressed nude mice was used. The tumor was induced by

subcutaneous injection of 7.5 x 106 Mia-PaCa-2 cells per mouse. TGM4 or TGM5 were

administrated orally and daily at a dose of 1000 mg/kg of body weight. Control mice

received nothing or gemcitabine 100 mg/kg i.p. twice a week, the standard of care for

the treatment of pancreatic cancer. The experiment lasted for 43 days, during which the

tumor volume was measured.

Figure 16. Representative images of the size of the pancreatic cancer xenograft in nude mice after

43 days of treatment. 7.5 million cells of the cell line Mia-PaCa-2 were injected subcutaneously in

Page 39: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

39

mice immunosuppressed NUDE Swiss Crl: NU (Ico) -Foxn1nu. (A) control mouse non-treated. (B)

mouse treated by intraperitoneal injection with Gemcitabine 100 mg/kg twice a week. (C) mouse

treated orally with 1000 mg/kg of TGM4. (D) mouse treated orally with 1000 mg/kg of TGM5.

The results obtained were visually appreciable at least on the TGM4 treated mice (Figure

16), and their tumors volume was significantly lower than control mice after 43 days of

treatment (Figure 17). The evolution of tumor size throughout the treatment time showed

that from day 12 of treatment, the tumors development of the treated and untreated

mice was different, being the control animal’s tumors always higher than those treated

with TGM4. The effect of the TGM5 was less potent than TGM4 but antitumor activity

was also observed (Figure 17 top). At the end of the treatment, a significant reduction in

tumor volumes of the TGM4 treated animals compared to control animals (Figure 18

bottom) was observed.

Page 40: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

40

Mia-Paca-2 xenograft volume at day 43

Contr

ol

Gem

cita

bine

(100

mg/k

g x2/

wee

k)

TGM

4 (1

000

mg/k

g day

)

TGM

5 (1

000

mg/k

g day

)

0

500

1000

1500

n=9

n=3

n=7

n=8

***

Tu

mo

r vo

lum

e (

%)

Figure 17. Effect of TGM4 / TGM5 in xenograft model of pancreatic cancer (Mia-PaCa-2). 7.5 x 106

Mia-PaCa-2 were injected subcutaneously in immunosupressed NUDE Swiss Crl: NU (Ico) -

Foxn1nu mice. Control animals received nothing or subcutaneous injections of 100 mg/kg of

Gemcitabine twice a week. The treated animals received through cannulation 1000 mg/Kg of

TGM4 or TGM5. (Top) Changes in the tumors volumes through 43 days of treatment represented

as the percentage. (Bottom) Percentage of tumors volumes at day 43 of treatment each value

represents the average of the percentages relative to day 0 (taken as 100%). non-parametric

Mann-Whitney test, (*p < 0.05; **p < 0.01).

The lipid compound TGM4 has shown to be at least as efficient as the actual standard of

care for the pancreatic cancer, the Gemcitabine.

4.4 Effect of the TGM4 on the cell cycle.

To establish the basis for the study of TGM4 effect in cell proliferation, first, PANC-1 cells

were treated with TGM4 and the cell cycle was analyzed. Two studies were conducted to

observe how the effect varies in relation to different doses and treatment times.

To study the dose-dependent effects, PANC-1 cells were treated at two different

concentrations, 20 and 40 µM for 48 hours (Figure 18). The results shows a clear dose-

dependency of the TGM4-induced effects. TGM4 treatment at both concentrations

produced a significant increase in the percentage of cells in sub-G1 phase (corresponding

to dead cells) followed by a decrease in the percentage of cells in G2/M phase and an

increase of the percentage of cells in the S phase.

Page 41: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

41

Sub-G

1

G0/

G1 S

G2/

M

0

15

30

45

60

Control

TGM4 20 µM

TGM4 40 µM

48hrs

PANC-1

* *

*

**

Cell

perc

en

tag

e (

%)

Figure 18. Effect of the concentration of TGM4 on the cell cycle in PANC-1 cell line. Cells were

treated with TGM4 at two concentrations (20 and 40 µM) for 48 hours and the cell cycle phases

populations were determined. Analysis in percentage of events in the different cell cycle phases

(sub-G1, G0/G1, S and G2/M) .Data represented as mean +- SEM (n=2), Statistical significance was

determined by unpaired two-tailed Student's t test (*P < 0.05)

In other set of experiments, PANC-1 cells were treated at a single concentration, 30 µM,

for 6, 12, 24 and 48 hours, to study the time-dependent effect (Figure 19). As control

treatment the same cells were treated with vehicle for the same time. The results show

a significant increase in the percentage of cells in sub-G1 phase with the treatment of

TGM4, associated to a state of cell death. This increase is accompanied by a significant

decrease of the percentage of cells in G0/G1 phase. The remaining phases were

unaffected by treatment TGM4 at the times studied, but the time-dependence effect and

cell death induction by TGM4 are confirmed.

Page 42: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

42

PANC-1

Sub-G

1

G0/

G1 S

G2/

M

Sub-G

1

G0/

G1 S

G2/

M

Sub-G

1

G0/

G1 S

G2/

M

Sub-G

1

G0/

G1 S

G2/

M

0

15

30

45

60

Control

TGM4 30 µM

6 hrs 12 hrs 48 hrs24 hrs

***

*

**

**

Cell

perc

en

tag

e (

%)

Figure 19. Effect of the time treated with TGM4 on the cell cycle in PANC-1 cell line. Cells were

treated with TGM4 30 µM for 6, 12, 24 and 48 hours and analyzed by flow cytometry to determine

the different cell cycle phases populations. Analysis in percentage of events in the different cell

cycle phases (sub-G1, G0/G1, S and G2/M). Data represented as mean +- SEM (n=4), Statistical

significance was determined by unpaired two-tailed Student's t test (*P < 0.05; ***P < 0.001).

Another confirmation of the results shown in the Figure 18 and 19 is the picture obtained

by microscopy of TGM4-treated PANC-1 cells (Figure 20), where the number of cells is

clearly reduced and the morphology is altered in a way that the cells seems to be

damaged and dying, as can be observed for the round shape and the presence of vesicles

in cytoplasm.

Figure 20. Representative pictures (400x) of PANC-1 cells with or without incubate with TGM4 30

µM for 48 h. The reduction of the number of cells and the loose of their epithelial morphology

can be observed in the TGM4-treated cells. No apoptotic bodies or blebbing (characteristic of

apoptotic cells) were observed.

Page 43: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

43

4.5 Finding the type of cell death and related proteins behind the TGM4 effect.

In order to know the type of cell death produced by TGM4 treatment and how affects

some of the main oncogenic signaling pathways, protein studies were performed trough

the classic Western Blot technique. PANC-1 cells were treated with TGM4 at a

concentration of 30 µM with different treatment times of 6, 12 and 24 hours. Then, they

were analyzed to determine the protein levels or phosphorylation state of key proteins

in stress and cell death processes, such as apoptosis, endoplasmic reticulum stress and

autophagy, as well as, proliferation induction.

Figure 21. TGM4 effect on different proteins studied. PANC-1 cells were treated with TGM4 30 µM

at different times (6, 12 and 24 hours), lysed, and an immunoblot was performed. In this

representative immunoblot it shows how the TGM4 affects proteins involved in reticular stress

and autophagy (BIP, CHOP and LC3B), the apoptosis reporter protein PARP, and to the protein

used as loading control α-tubulin.

As observed previously by flow cytometry and XTT assay, TGM4 induced cell death in

TGM4, based in that, the levels of key protein markers for apoptosis and autophagy were

studied (Figure 21). The fragmentation of PARP, typical indicator of apoptosis, was not

observed, but there was a clear downregulation of the full form since the 6 hours of

treatment. On the other hand, the increase of BIP indicates reticular stress on the treated

cells. Besides, the stress inducible CHOP was clearly expressed at 12 and 24 hours, in a

time- and dose-dependent manner. In a similar way, the bottom fragment of LC3B (LC3B-

II) is upregulated upon TGM4 treatment. Knowing that LC3B-II is an essential protein for

the formation of autophagosomes and so for autophagy, TGM4 can be considered an

inducer of autophagy in PANC-1 cells.

Page 44: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

44

After determining that PANC-1 cells die through autophagy, and taking into account that

TGM4 modified other cell cycle phases populations, the protein levels alteration of critical

proteins for the right cellular proliferation were studied to try determining the sequence

of the events (Figure 22). For example, a downregulation of the levels of DHFR,

dihydrofolate reductase, an enzyme necessary for the synthesis of DNA was observed.

Two proteins involved in the regulation of the cell cycle were studied, Cyclin D3 and p19.

The downregulation of Cyclin D3 is remarkable from the 12 hours, justifying the decrease

of cells in G1/G0 phase, but p19 remained unaltered, corresponding with the cell death

instead of cell cycle arrest. Finally, as important oncogenic signaling pathway the

PI3K/AKT pathway, which contributes to proliferation and survival, was studied through

the protein AKT, and changes in its regulation by phosphorylation and its total protein

were observed, but not a clear upregulation or downregulation. Moreover, the mitogen-

activated protein kinases (MAPK) pathway, also involved in the regulation of proliferation,

Figure 22. TGM4 effect on different proteins studied. PANC-1 cells were treated with TGM4 30 uM

at different times (6, 12 and 24 hours), lysed, and an immunoblot was performed. In this

representative immunoblot it shows how the TGM4 affects proteins of pathways involved in

oncogenesis (Jun, AKT and ERK), regulatory proteins of the cell cycle (CD3 and p19), protein

required for DNA synthesis DHFR, and to the protein used as loading control α-tubulin.

Page 45: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

45

was studied through the protein ERK, and as in the AKT case, both regulation and total

protein were affected in different forms depending of the time. Interestingly, c-jun is a

proto-oncogene involved in proliferation and apoptosis, and its activity was studied

through the protein Jun, in this case only its phosphorylation state was affected by TGM4

treatment indicating an inhibition, being the levels of total Jun unaltered by the TGM4.

4.6 Lipid alterations produced by the treatment with TGM4

When working with lipid compounds that are intended to intercalate into the lipid

membrane and change its structure to produce effects in cell proliferation and survival,

the study of specific alterations produced in the different cellular lipids becomes

important. For this, PANC-1 cells were treated at two concentrations of TGM4 (20 and 30

µM) for 12 or 48 hours, then a TLC was carried to separate the major phospholipids

present in the whole cell.

Page 46: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

46

Figure 23 Thin-layer chromatography quantification after treat PANC-1 cells with TGM4 30 µM for

12h (Top) or 48h (Bottom). Data are presented as mean ± SEM. Statistical significance was

determined by unpaired two-tailed Student's t test (*p < 0.05; **p < 0.01). CLP: cardiolipin; PE:

phosphatidylethanolamine; PI: phosphatidylinositol; PS: phosphatidylserine; PC:

phosphatidylcholine; SM: Sphingomyelin.

A significant increase of cardiolipin was found after 12 hours of treatment with TGM4

(Figure 23 Top). Intriguingly, this increase disappeared after 48 hours of treatment (Figure

23 Bottom). There was also a little but significant downregulation of Phosphatidylcholine

after 12 hours of incubation with TGM4 (Figure 23 Top). The rest of the studied lipids

remained constant for all the conditions.

Page 47: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

47

5. Discussion

5.1 The TGMs, a novel library of compounds, are effective antiproliferative drugs.

The pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death

in the United States, sixth in Europe, despite of the relative short incidence of 10-

12:100.00.

The high mortality due to their aggressive nature, the difficulty of an early detection and

thus its high metastatic rate, besides the lack of effective treatments for control the tumor

if surgical resection fails, make of this cancer one of the cancers with more clinical

significance nowadays. Of all pancreatic tumor cell lines we chose working with Mia-

PaCa-2 for growth studies and with PANC-1 line for molecular studies, because the last

line was obtained from ductal cells and it represents better at molecular level the cancer

that mostly affects humans.

The library of compounds named TGMs are rationally designed from different natural

triglycerides. The hypothesis of this study is that all the molecules studied due to their

lipid character will act on the lipid membrane, regulating the composition and membrane

fluidity. Once in the membrane they will affect the interaction and activity of peripheral

membrane proteins involved in several signaling pathways that are eventually related to

cellular membrane and the regulation of vital cellular processes for oncogenesis. In

summary, the changes produced by the action of these molecules will cause several

changes in cell signaling pathways that ultimately end up producing changes in the

processes of cell proliferation and survival.

In vitro studies carried out for this master thesis showed that the use of the molecules

tested decreases the number of pancreatic tumor cells, and that the number of cells

decreases in proportion to the concentration and time.

It is noteworthy that the differences in the IC50s of the molecules are not significantly

different and generally most of them are considered to show good antitumor potency.

The most potent molecules, those who have a lower IC50 were TGM5, TGM12 and

TGM25, but for selecting a molecule to develop and study deeply there are other factors

to take into account as its structure. That is the reason why at the end TGM4 was

highlighted, the free fatty acids that are part of its structure are promising antitumor

candidates by themselves.

Another factor to consider evaluating the antitumor activity of a compound is its efficacy.

The potency of a compound is measured mainly from its IC50, so a compound has with

Page 48: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

48

a lower IC50 has greater potency. The efficacy here is reflected by the ability of the

compound to kill, its maximal therapeutic effect. Most of the TGMs showed to kill all the

cells of the culture.

The benefits associated with great antitumor efficacy and potency (low concentrations

needed) are remarkable, because at low concentration decrease the possibility that

appear secondary or unwanted side effects. For that reason we chose TGM5 to test its

effect on in vivo models, together with the TGM4 that we chose for structural reasons.

The study of the mixed TGMs, though very interesting, was postponed for future

experiments.

Even after checking TGM4 and TGM5 lack of toxicity evaluating the survival, no significant

body weight loose and no behavioral effect of mice after their administration, it would

be necessary to test their toxicity and effect in some non-tumor cell lines, for example

MRC-5 lung human fibroblasts, that would allow to determine if we are within the range

of normal cell toxicity. As they has shown to be non-toxic for mice, we hypothesize that

they will have a greater effect on cancer cells than in normal cells, which is a must in

therapies against these diseases. Specific drugs are sought to make these effects will

occur only in cancer cells, thus preventing a large number of unwanted side effects.

For future studies there are remaining few molecules of the TGMs library that, for time

reasons, does not have been tested in this master thesis and based on the results of the

rest of compounds we hypothesize that they will work fine as antiproliferative drugs.

On the other hand there are more pancreatic cancer cell lines and lines from other kind

of important cancers in which the molecules (or at least some of them) should be tested.

If the molecules do not works fine on other cancer cell lines there should be some

specifically characteristic of the pancreatic cancer cells that make them sensitive to the

molecules. If the tested molecules work on several different cancer cell lines will mean its

effect is produced disrupting some essential oncogenic step. Both possibilities will offer

important data to understand how the TGMs works.

5.2 The TGM4 is an effective antitumor drug, tested on Mia-PaCa-2 xenograft in

vivo models of pancreatic cancer.

In vivo studies in nude immunodepressed mice confirmed that the antitumor effect of

the molecule TGM4 against pancreatic cancer. Unfortunately, TGM5 did not have

significant antitumor activity in these studies. Regarding the effect produced by the

TGM4 and the treating conditions, there are remarkable points.

Page 49: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

49

When an animal or a human has to be treated with any drug due to cancer disease, the

toxicity of the compound is the factor to take into account for deciding the most effective

dose but with the minimum side effects. From the toxicity studies the dose of 1000 mg/kg

was chosen without any adverse effect that could make us consider that TGM4 could be

dangerous for a potential future patient.

As shown in Figure 17 the effect of the TGM4 at a daily oral dose of 1000 mg/kg is very

similar to that produced by the drug of reference gemcitabine at a dose of 100 mg / kg

given to the mice by intraperitoneal injection twice a week. When considering a drug for

antitumor therapy in humans are taken into account several factors, but two essential are

potency and toxicity. The fact that the effect has been the same even though the TGM4

dose was much higher may suggest that the TGM4 is less effective for the treatment of

pancreatic cancer than gemcitabine, but not necessarily. The possibility of such a high

dose without toxicity effects show that the TGM4 is an antitumor molecule with a low

toxicity profile, which provides a greater therapeutic window, ie, can be administered at

higher doses without the fear of producing the known side effects of most

chemotherapeutic drugs, which may range from mild to very severe.

Furthermore, the fact that the TGM4 can be administered orally maintaining its antitumor

effect provides information about its absorption and distribution. For a triglyceride orally

ingested to reach its destination, it must be absorbed in the intestine. Triglycerides form

a fat emulsion by the action of bile salts and these are processed by digestive lipases that

cleave fatty acids and glycerol to be absorbed separately by intestinal epithelial cells.

Within these enterocytes, triglycerides are reformed and transported, together with

proteins, lymph circulation and blood (Trauner et al. 2010). Once in the blood, triglyceride

can reach the cells that form the tumor, where lipoproteins lipases from the endothelial

cells cleave the triglycerides again and allow the fatty acids and glycerol enter the cell by

the vascular endothelium, penetrating in the tumor cell wherein the fatty acids will

produce their antiproliferative effect.

Oral administration is an advantage over intravenous administration, because apart from

allowing patients avoid continued injections, it has a number of benefits over other

routes of administration. Oral delivery is suitable for the vast majority of patients

regardless of age or physical condition, is a natural way and completely painless. In

addition there are several possible formats for oral administration, whether tablets,

capsules, syrups, emulsions...and these do not require medical attention for their

administration. Finally, we consider that the most important point is that oral

administration is associated with less toxicity due to the fact that the absorption is

prolonged and sustained, allowing better tolerance to side effects of the drug.

Page 50: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

50

Additionally and especially in pancreatic cancer, tumors eventually produce resistance to

most drugs after continued treatment time. Thus, for example, for pancreatic cancer the

only therapy that is considered to have curative potential is resection of the affected area

of the pancreas, and other treatments are palliative, because usually the treatment does

not remove 100% of the tumor cells before they produce resistance. The TGM4 by its

lipid nature would be included within the known Membrane-Lipid Therapy, which affect

not only a unique protein as current drugs do. TGM4 would affect current up to several

signaling pathways that are initiated in the membrane. It has been observed the ease of

tumor cells for produce metabolic bypasses so they avoid the proteins that are used as

targets for anti-tumor cell growth. These bypasses would be much more difficult to

perform if several signaling pathways in the initial point are affected, so it is hypothesized

that the development of resistance to the molecules acting through MLT would be much

lower than in conventional chemotherapy.

In the study conducted, a clear and significant reduction of the tumor caused by the

effect of TGM4 was observed. We should repeat the experiments to confirm the observed

results. Once confirmed the effect of the TGM4 in vivo, this could be an effective therapy

against pancreatic cancer. Currently and increasingly, it is pointing to the use of

customized and combined with different chemotherapy drugs that allow greater control

over the different signaling pathways targeted therapies. This combination with other

chemotherapy drugs would allow to reduce the dose of chemotherapy and thus reduce

the high toxicity that often appear in patients. Ease of administration and low toxicity of

the TGM4 make this molecule an ideal candidate to offer an alternative as monotherapy

or in combination in patients with pancreatic cancer.

5.3 TGM4 produces an antiproliferative effect and death by autophagy in cancer

cells PANC-1.

The study of the TGM4 mechanism of action of revealed that induces cell death. The cell

cycle analysis of TGM4-treated cells with this drug for 6, 12, 24 and 48 hours showed an

increase of cell death at concentrations near to its IC50 (Figure 19). The cell death was

confirmed by an increase in the percentage of cells found in the sub-G1 phase. As a result

of this increase, a significant decrease in the percentage of cells in the G0 / G1 phase of

the cell cycle.

However, in the studies of dose-dependency (Figure 18) the increase in the percentage

of cells found in the sub-G1 phase was followed by a decrease of the cell population in

the S and G2/M phases, not in in the G0/G1 phase as in figure 19. This studies have to

be repeated in order to confirm the results, but a priori, by the statistical significance of

Page 51: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

51

the experiments, we believe that the decrease of the cell population in G0/G1 phase is

more interesting and significant.

The findings in the cell cycle may be related to the alterations found in some cell cycle

regulatory proteins. In the protein studies, it was found that the treatment with TGM4

produced a drop in the cyclin D3 levels. Cyclin D3 acts as a regulatory subunit of a

complex formed with Cdk4 or Cdk6 and its activity is required for the G1/S cell cycle. As

this is a protein necessary for the transition or regulation from the G0/G1 phase to the S

phase and the TGM4 produced a decrease in the population of cells in the G0 / G1 phase,

reduction of this protein levels is expected and supports the results obtained in the flow

cytometry. On the other hand, p19 protein has-been shown to form a stable complex

with CDK4 or CDK6, preventing the activation of the CDK kinases, functioning as a cell

growth regulator that controls the G1 cell cycle progression and induces the arrest in

that cycle. This protein levels were not affected by the treatment with TGM4, in

accordance with the drop of the cell number in G0/G1 and cell death instead of cell cycle

arrest. Other protein important for the cell proliferation included in this study is DHFR.

This protein is required for DNA synthesis and therefore to cell replication, especially if

this is accelerated as in cancer. TGM4 decreases expression of the DHFR protein, which

hampers DNA replication to cancer cells and thus cell replication. Some drugs such as

methotrexate and have as target the expression of DHFR protein but are highly toxic.

Part of the TGM4 effect may be due to its effect in reducing the expression of DHFR.

Other lipid compounds developed in this group have shown that DHFR downregulation

is key for their effect (Lladó et al. 2009)

The study of cell proliferation signaling pathways indicates that the TGM4 affects the

regulation by phosphorylation of the studied proteins AKT, ERK, and c-jun. Unexpectedly,

we found that in the AKT and ERK cases, the stability or expression of the protein was

affected also, because an alteration in the total levels of these proteins (not only in the

phosphorylated protein) were found.

Specifically, TGM4 affects the PI3K/AKT and ERK pathways, showing variations in the

phosphorylation and total levels of AKT and ERK proteins. However, the effect on them

is variable, not a clear dose or time dependent upregulation or downregulation, one

possibility is that AKT is being altered in response of the pathways directly modified by

TGM4, for example, AKT upregulation at 6hrs could be due to an attempt to

counteracting the antiproliferative effects and stress signals of other pathways, such as

JNK pathway or BIP/CHOP induction. This hypothesis would need more exploration and

could be accepted and discarded after we establish a timeline. In the case of c-jun we

found the TGM4 reduced the levels of phosphorylated c-jun, but TGM4 did not affected

the total levels of Jun. The alterations of this pathways are highly involved in the

Page 52: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

52

regulation of proliferation and expected when working with an antiproliferative drug.

These results may raise suspicion of the cellular stress that TGM4 produces, a fact that

can be confirmed by the overexpression of the protein BIP treatment cells are found. BIP

is a protein which recognizes misfolded proteins, and protein subunits that are not

assembled into their final oligomer complexes, so the presence of BIP is associated with

a reticular stress.

On the other hand we sought to discover what type of cell death is produced by the

TGM4, apoptosis was almost ruled out from the start by the non-presence of apoptotic

bodies in the cultivation nor evidence in cell morphology that indicate apoptosis after

treatment with TGM4 (Figure 20). To discard apoptosis, PARP protein fragmentation was

analyzed because it is considered an indicator of apoptosis. As, fragmentation of PARP

was not observed, the apoptosis via caspases cascade was discarded. However, in

caspase independent apoptosis, a downregulation of full length PARP (116 kDa) is

produced. Since in our experiments this downregulation of the full PARP is observed, it

would be interesting to investigate. Because autophagy and apoptosis are not exclusive.

When analyzing autophagy related proteins, positive results were obtained. Autophagy

is an intracellular bulk degradation system, through which a portion of the cytoplasm is

delivered to lysosomes to be degraded. Aggregation and posttranscriptional

modification of microtubule-associated protein light chain 3 (LC3), a mammalian

homolog of yeast Atg8, has-been used as a specific marker to monitor autophagy (Kuma

et al. 2007). LC3B is first cleaved at the carboxy terminus immediately following synthesis

to yield a cytosolic form LC3B-I. During autophagy LC3B-I is converted to LC3B-II through

lipidation by an ubiquitin-like system involving Apg7 and Apg3 that allows for LC3 to

become associated with autaphagic vesicles (Kabeya et al. 2000; He et al. 2003; Tanida et

al. 2004; Wu et al. 2006; Ichimura et al. 2000). Thus, the presence of LC3 in

autophagosomes as well as the conversion of LC3 to the lower migrating form LC3B-II

have been used as indicators of autophagy. The marked overexpression of LC3B-II after

the treatment with TGM4 indicates that autophagy is triggered in treated cells.

One candidate to be the link between the ER stress and autophagy is CHOP, it encodes

a ubiquitous transcription factor that is one of the most important components in the

network of stress-inducible transcription. In particularly, this factor is known to mediate

cell death in response to stress (B´chir et al. 2014). CHOP promotes IRE1α and autophagy

especially in ER stress conditions. (Shimodaira et al. 2014). Increasing levels of CHOP were

found in the TGM4 treated cells and this corresponds with an increasing of the protein

BIP (associated with reticular stress) and increased LC3B-II (associated with autophagy).

As a part of his mechanism of action, we suggest that TGM4 produces a reticular stress

Page 53: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

53

in the cells, being CHOP one of the responsible of promoting autophagy given the

situation of reticular stress caused by TGM4 and confirmed by overexpression of BIP.

All work performed in this master thesis is based on the theory that the compounds used,

for its lipid nature, will be inserted in the membrane and eventually leading to changes

in the activity of peripheral proteins membrane. The study of the alteration produced by

the TGM4 in the composition of the lipid membrane was limited to the phospholipids in

the cell whole extract, as only a reduced number of lipids were studied, among the

hundreds present in a cell. But despite this, a remarkable increasing of the cardiolipin

levels and a little but significant decrease in the phosphatydilcholine levels was found.

Further study of more lipids is needed to establish the relationship between the

mechanism of action showed by TGM4 and the alteration produced in membrane lipids

levels or composition. Indeed, recently, our group has published that TGM1 treatment (a

mimetic triglyceride derived from triolein, not included in this master thesis) in lung

cancer cells modifies the levels of neutral lipids (DAG, TAG and free fatty acids) what

induces an increase of ROS production in the cell, triggering autophagy cell death

(Guardiola-Serrano et al. 2015). Besides, cardiolipin is the most abundant lipid in the

mitochondria and the alteration of its levels/localization could induce mitophagy, other

possible cause of cell death that could be explored.

Generally, more studies are needed to clarify the sequence of events leading to the

confirmed cell death. The possibilities of membrane-lipid therapy are high because

altering the lipid membrane, we find not only a single change to the level of a target

protein as with current treatments. By targeting the membrane, restoring the lipid levels

and distribution to a normal state, a single compound can produce countless changes in

signaling pathways and thus in biological processes associated with proliferation or

apoptosis, which may be decisive for the regulation of oncogenic processes.

Page 54: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

54

6. Conclusions

The main conclusions of this master thesis are:

1. The studies performed for this master thesis highlight the idea that the Membrane-

Lipid Therapy can be effective in the treatment of an important disease as cancer

2. The TGMs library exerts a remarkable antiproliferative effect on pancreatic cancer cells

Mia-PaCa-2.

3. TGM4 causes cell death by reticular stress followed by autophagy in PANC-1 cells.

4. The TGM4 affects important signaling pathways, like the SAPK/JNK/c-jun pathway, and

other studied proteins. More studies are needed to clarify the sequence of events leading

to cell death.

5. Studies in vivo: chronic administration of TGM4 reduces the progression of tumors in

nude mice inoculated with Mia-PaCa-2 cells.

Page 55: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

55

7. References

Barbacci, E.G. et al., 2003. The biological and biochemical effects of CP-654577, a selective

erbB2 kinase inhibitor, on human breast cancer cells. Cancer research, 63(15),

pp.4450–9. Available at: http://www.ncbi.nlm.nih.gov/pubmed/12907618

Birklé, S. et al., Role of tumor-associated gangliosides in cancer progression. Biochimie,

85(3-4), pp.455–63. Available at: http://www.ncbi.nlm.nih.gov/pubmed/12770784

Burris, H. 3rd et al., 1997. Improvements in survival and clinical benefit with gemcitabine

as first- line therapy for patients with advanced pancreas cancer: a randomized trial.

J. Clin. Oncol., 15(6), pp.2403–2413. Available at:

http://jco.ascopubs.org/content/15/6/2403.short

Chen, N. & Karantza, V., 2011. Autophagy as a therapeutic target in cancer. Cancer

Biology & Therapy. Available at:

http://www.tandfonline.com/doi/abs/10.4161/cbt.11.2.14622#.VexJP_ntmko

Cheung, Z.H. & Ip, N.Y., 2011. Autophagy deregulation in neurodegenerative diseases -

recent advances and future perspectives. Journal of neurochemistry, 118(3), pp.317–

25. Available at: http://www.ncbi.nlm.nih.gov/pubmed/21599666

Clerc, P. et al., 1991. Lipid-dependent Proliferation of Pancreatic Cancer Cell Lines Lipid-

dependent Proliferation of Pancreatic Cancer Cell Lines1. , pp.3633–3638.

Cullis, P.R. & De Kruijff, B., 1979. Lipid polymorphism and the functional roles of lipids in

biological membranes. Biochimica et Biophysica Acta (BBA) - Reviews on

Biomembranes, 559(4), pp.399–420. Available at:

http://www.sciencedirect.com/science/article/pii/0304415779900121

Dekoj, T. et al., 2007. G2/M cell-cycle arrest and apoptosis by n-3 fatty acids in a

pancreatic cancer model. The Journal of surgical research, 139(1), pp.106–12.

Available at: http://www.ncbi.nlm.nih.gov/pubmed/17291531Easton, J.B. &

Houghton, P.J., 2006. mTOR and cancer therapy. Oncogene, 25(48), pp.6436–46.

Available at: http://dx.doi.org/10.1038/sj.onc.1209886

Eberhart, K., Oral, O. & Gozuacik, D., 2014. Induction of Autophagic Cell Death by

Anticancer Agents, Elsevier Inc. Available at:

http://linkinghub.elsevier.com/retrieve/pii/B9780124055308000133.

Engelman, D.M., 2005. Membranes are more mosaic than fluid. Nature, 438(7068),

pp.578–80. Available at: http://dx.doi.org/10.1038/nature04394

Escribá, P. V et al., 2008. Membranes: a meeting point for lipids, proteins and therapies.

Journal of cellular and molecular medicine, 12(3), pp.829–75. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/18266954

Page 56: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

56

Escriba, P. V. et al., 1997. Role of lipid polymorphism in G protein-membrane interactions:

Nonlamellar-prone phospholipids and peripheral protein binding to membranes.

Proceedings of the National Academy of Sciences, 94(21), pp.11375–11380.

Available at: http://www.pnas.org/content/94/21/11375.abstract

Escriba, P. V. et al., 1990. Role of membrane lipids in the interaction of daunomycin with

plasma membranes from tumor cells: implications in drug-resistance phenomena.

Biochemistry, 29(31), pp.7275–7282. Available at:

http://dx.doi.org/10.1021/bi00483a017

Everhart, J. & Wright, D., Diabetes mellitus as a risk factor for pancreatic cancer. A meta-

analysis. JAMA, 273(20), pp.1605–9. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/7745774

Falconer, J. et al., 1994. Effect of eicosapentaenoic acid and other fatty acids on the

growth in vitro of human pancreatic cancer cell lines. British Journal of Cancer, 69(5),

pp.826–832. Available at: http://www.nature.com/doifinder/10.1038/bjc.1994.161.

Fiorini, C. et al., 2015. Onconase induces autophagy sensitizing pancreatic cancer cells to

gemcitabine and activates Akt/mTOR pathway in a ROS-dependent manner.

Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 1853(3), pp.549–560.

Available at: http://linkinghub.elsevier.com/retrieve/pii/S0167488914004443.

Fukui, M. et al., 2013. EPA, an omega-3 fatty acid, induces apoptosis in human pancreatic

cancer cells: role of ROS accumulation, caspase-8 activation, and autophagy

induction. Journal of cellular biochemistry, 114(1), pp.192–203. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/22903547

Funderburk, S.F., Wang, Q.J. & Yue, Z., 2010. The Beclin 1-VPS34 complex--at the

crossroads of autophagy and beyond. Trends in cell biology, 20(6), pp.355–62.

Available at: http://www.sciencedirect.com/science/article/pii/S096289241000053X

Gapstur, S.M. et al., 2000. Abnormal glucose metabolism and pancreatic cancer mortality.

JAMA, 283(19), pp.2552–8. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/10815119

Gottlieb, R.A. & Carreira, R.S., 2010. Autophagy in health and disease. 5. Mitophagy as a

way of life. AJP: Cell Physiology, 299(2), pp.C203–C210. Available at:

http://ajpcell.physiology.org/content/299/2/C203.short

Guardiola-Serrano, F., Beteta-Göbel, R., Rodríguez-Lorca, R., Ibarguren, M., López, D. J.,

Terés, S., ... & Escribá, P. V. 2015. The Novel Anticancer Drug Hydroxytriolein Inhibits

Lung Cancer Cell Proliferation via a Protein Kinase Cα–and Extracellular Signal-

Regulated Kinase 1/2–Dependent Mechanism. Journal of Pharmacology and

Experimental Therapeutics, 354(2), 213-224.

Hanahan, D. & Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell,

144(5), pp.646–74. Available at: http://www.ncbi.nlm.nih.gov/pubmed/21376230

Page 57: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

57

Hanahan, D. & Weinberg, R.A., 2000. The Hallmarks of Cancer. Cell, 100(1), pp.57–70.

Available at: http://www.sciencedirect.com/science/article/pii/S0092867400816839

He, H. et al., 2003. Post-translational modifications of three members of the human

MAP1LC3 family and detection of a novel type of modification for MAP1LC3B. The

Journal of biological chemistry, 278(31), pp.29278–87. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/12740394

Hettmer, S., Ladisch, S. & Kaucic, K., 2005. Low complex ganglioside expression

characterizes human neuroblastoma cell lines. Cancer letters, 225(1), pp.141–9.

Available at:

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2866625&tool=pmcen

trez&rendertype=abstract

Hezel, A.F. et al., 2006. Genetics and biology of pancreatic ductal adenocarcinoma

Genetics and biology of pancreatic ductal adenocarcinoma. , 1, pp.1218–1249.

Ibarguren, M. et al., 2013. Partitioning of liquid-ordered/liquid-disordered membrane

microdomains induced by the fluidifying effect of 2-hydroxylated fatty acid

derivatives. Biochimica et biophysica acta, 1828(11), pp.2553–63. Available at:

http://www.sciencedirect.com/science/article/pii/S0005273613002034

Ichimura, Y. et al., 2000. A ubiquitin-like system mediates protein lipidation. Nature,

408(6811), pp.488–92. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/11100732

Igal, R.A., 2010. Stearoyl-CoA desaturase-1: a novel key player in the mechanisms of cell

proliferation, programmed cell death and transformation to cancer. Carcinogenesis,

31(9), pp.1509–15. Available at: http://www.ncbi.nlm.nih.gov/pubmed/20595235

Jemal, A. et al., Cancer statistics, 2009. CA: a cancer journal for clinicians, 59(4), pp.225–

49. Available at: http://www.ncbi.nlm.nih.gov/pubmed/19474385.

Kabeya, Y. et al., 2000. LC3, a mammalian homologue of yeast Apg8p, is localized in

autophagosome membranes after processing. The EMBO journal, 19(21), pp.5720–

8. Available at:

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=305793&tool=pmcentr

ez&rendertype=abstract

Kuhajda, F.P., 2006. Fatty acid synthase and cancer: new application of an old pathway.

Cancer research, 66(12), pp.5977–80. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/16778164

Lai, P. et al., 1996. Cell cycle arrest and induction of apoptosis in pancreatic cancer cells

exposed to eicosapentaenoic acid in vitro. British Journal of Cancer, 74(9), pp.1375–

1383. Available at: http://www.nature.com/doifinder/10.1038/bjc.1996.552.

Page 58: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

58

Levine, B., Mizushima, N. & Virgin, H.W., 2011. Autophagy in immunity and inflammation.

Nature, 469(7330), pp.323–35. Available at: http://dx.doi.org/10.1038/nature09782

Lingwood, D. & Simons, K., 2010. Lipid rafts as a membrane-organizing principle. Science

(New York, N.Y.), 327(5961), pp.46–50. Available at:

http://www.sciencemag.org/content/327/5961/46.short

Lladó, V. et al., 2009. Pivotal role of dihydrofolate reductase knockdown in the anticancer

activity of 2-hydroxyoleic acid. Proceedings of the National Academy of Sciences of

the United States of America, 106(33), pp.13754–8. Available at:

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2720411&tool=pmcen

trez&rendertype=abstract

Lladó, V. et al., 2014. Regulation of the cancer cell membrane lipid composition by

NaCHOleate: Effects on cell signaling and therapeutical relevance in glioma.

Biochimica et Biophysica Acta - Biomembranes, 1838(6), pp.1619–1627. Available at:

http://dx.doi.org/10.1016/j.bbamem.2014.01.027.

LOWRY, O.H. et al., 1951. Protein measurement with the Folin phenol reagent. The Journal

of biological chemistry, 193(1), pp.265–75. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/14907713

MacKenzie, K. et al., 2001. Effect of eicosapentaenoic acid and other fatty acids on the

growth in vitro of human pancreatic cancer cell lines. BMJ. Available at:

http://www.research.ed.ac.uk/portal/files/11824718/Is_voice_therapy_an_effective_

treatment_for_dysphonia.pdf

McMullen, T.P.W., Lewis, R.N.A.H. & McElhaney, R.N., 2004. Cholesterol–phospholipid

interactions, the liquid-ordered phase and lipid rafts in model and biological

membranes. Current Opinion in Colloid & Interface Science, 8(6), pp.459–468.

Available at: http://www.sciencedirect.com/science/article/pii/S1359029404000081

Van Meer, G. & de Kroon, A.I.P.M., 2011. Lipid map of the mammalian cell. Journal of cell

science, 124(Pt 1), pp.5–8. Available at:

http://jcs.biologists.org/content/124/1/5.short

Michaud, C.M., Murray, C.J. & Bloom, B.R., 2001. Burden of disease--implications for

future research. JAMA, 285(5), pp.535–9. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/11176854

Mikirova, N. et al., 2004. Erythrocyte membrane fatty acid composition in cancer patients.

Puerto Rico health sciences journal, 23(2), pp.107–13. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/15377058

Mirzoeva, O.K. et al., 2011. Autophagy suppression promotes apoptotic cell death in

response to inhibition of the PI3K-mTOR pathway in pancreatic adenocarcinoma.

Journal of Molecular Medicine, 89(9), pp.877–889.

Page 59: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

59

Mizushima, N. & Levine, B., 2010. Autophagy in mammalian development and

differentiation. Nature cell biology, 12(9), pp.823–30. Available at:

http://www.nature.com/ncb/journal/v12/n9/full/ncb0910-823.html%3Fmessage-

global%3Dremove

Morad, S. a F. et al., 2013. Potential role of acid ceramidase in conversion of cytostatic to

cytotoxic end-point in pancreatic cancer cells. Cancer chemotherapy and

pharmacology, 71(3), pp.635–45. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/23263160

Omura, N. & Goggins, M., 2009. Epigenetics and epigenetic alterations in pancreatic

cancer. International Journal of Clinical and Experimental Pathology, 2(4), pp.310–

326.

Ottenhof, N. a et al., 2011. Molecular characteristics of pancreatic ductal adenocarcinoma.

Pathology research international, 2011, p.620601.

Pike, L.J., 2006. Rafts defined: a report on the Keystone Symposium on Lipid Rafts and

Cell Function. Journal of lipid research, 47(7), pp.1597–8. Available at:

http://www.jlr.org/content/47/7/1597.short

Rabinowitz, J.D. & White, E., 2010. Autophagy and metabolism. Science (New York, N.Y.),

330(6009), pp.1344–8. Available at:

http://www.sciencemag.org/content/330/6009/1344.short

Rashid, A. et al., 1997. Elevated expression of fatty acid synthase and fatty acid synthetic

activity in colorectal neoplasia. The American journal of pathology, 150(1), pp.201–

8. Available at:

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1858511&tool=pmcen

trez&rendertype=abstract

Riboni, L. et al., 2002. Ceramide levels are inversely associated with malignant progression

of human glial tumors. Glia, 39(2), pp.105–13. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/12112362

Rubinsztein, D.C., Mariño, G. & Kroemer, G., 2011. Autophagy and aging. Cell, 146(5),

pp.682–95. Available at:

http://www.sciencedirect.com/science/article/pii/S0092867411008282

Shimodaira, Y. et al., 2014. Modulation of endoplasmic reticulum (ER) stress-induced

autophagy by C/EBP homologous protein (CHOP) and inositol-requiring enzyme 1α

(IRE1α) in human colon cancer cells. Biochemical and biophysical research

communications, 445(2), pp.524–33. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/24565834

Singer, S. J., & Nicolson, G. L. 1972. The fluid mosaic model of the structure of cell

membranes. Day and Good Membranes and viruses in immunopathology, 7-47.

Page 60: Javier Fernández - Study of the effects of mimetic triglycerides on human pancreatic cancer - Master thesis

60

Stolzenberg-Solomon, R.Z. et al., 2005. Insulin, glucose, insulin resistance, and pancreatic

cancer in male smokers. JAMA, 294(22), pp.2872–8. Available at:

http://jama.jamanetwork.com/article.aspx?articleid=202010 [Accessed September

6, 2015].

Swinnen, J. V et al., 2000. Stimulation of tumor-associated fatty acid synthase expression

by growth factor activation of the sterol regulatory element-binding protein

pathway. Oncogene, 19(45), pp.5173–81. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/11064454

Tanida, I., Ueno, T. & Kominami, E., 2004. Human light chain 3/MAP1LC3B is cleaved at

its carboxyl-terminal Met121 to expose Gly120 for lipidation and targeting to

autophagosomal membranes. The Journal of biological chemistry, 279(46),

pp.47704–10. Available at: http://www.ncbi.nlm.nih.gov/pubmed/15355958

Yeo, T.P. et al. 2002. Pancreatic cancer, Current Problems in Cancer, 26(4):176–275.

Vigh, L. et al., 2005. The significance of lipid composition for membrane activity: new

concepts and ways of assessing function. Progress in lipid research, 44(5), pp.303–

44. Available at:

http://www.sciencedirect.com/science/article/pii/S0163782705000305

Vögler, O. et al., 2004. The Gbetagamma dimer drives the interaction of heterotrimeric Gi

proteins with nonlamellar membrane structures. The Journal of biological chemistry,

279(35), pp.36540–5. Available at: http://www.ncbi.nlm.nih.gov/pubmed/15231827

Wiederschain, G.Y., 2006. The structure of biological membranes. Biochemistry (Moscow),

71(3), pp.345–345. Available at:

http://link.springer.com/10.1134/S0006297906030175

Wu, J. et al., 2006. Molecular cloning and characterization of rat LC3A and LC3B--two

novel markers of autophagosome. Biochemical and biophysical research

communications, 339(1), pp.437–42. Available at:

http://www.ncbi.nlm.nih.gov/pubmed/16300744.

Yang, Z.-Y. et al., 2013. Gemcitabine plus erlotinib for advanced pancreatic cancer: a

systematic review with meta-analysis. PloS one, 8(3), p.e57528. Available at:

http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0057528.