Você está na página 1de 23

Page | 1

1. Introduction
1.1 Genes and Cancer
Advances in science have improved our knowledge of the inner workings of cells, the basic
building blocks of the body. All living things are made of cells. Complex animals such as
humans have trillions of cells. Cells work together to form organs, such as the heart, liver,
and skin. Human bodies have several organ systems. Cancer begins when genes in a cell
become abnormal and the cell starts to grow and divide out of control.

1.1.1 Genes
Genes are pieces of DNA (deoxyribonucleic acid) inside each cell that tell the cell what to do
and when to grow and divide. Each gene is made up of a specific DNA sequence that
contains the code (the instructions) to make a certain protein, each of which has a specific job
or function in the body. Each human cell has about 25,000 genes. Most genes are contained
in chromosomes.
A chromosome is a long strand of DNA wrapped around a special protein called histone.
Most chromosomes contain many different genes. Most human cells contain 23 pairs of
chromosomes one pair of sex chromosomes (either XX in females or XY in males) plus 22
pairs of non-sex chromosomes called autosomes. Sperm and egg cells only contain half as
many chromosomes (23). Chromosomes are passed from parents to their children through
sperm and egg cells. One chromosome of each pair is inherited from the mother, and the
other comes from the father. This is why children look like their parents, and why they may
have a tendency to develop certain diseases that run in their families. A cell uses its genes
selectively; that is, it can turn on (or activate) the genes it needs at the right moment and turn
off other genes that it doesn't need. All the cells in the body (except egg and sperm) contain
the same genes. Turning on some genes and turning off others is how a cell becomes
specialized. That is how a cell becomes a muscle cell and not a bone cell, for example. Some
genes stay active all the time to make proteins needed for basic cell functions. Others shut
down when their job is finished and start again later if needed.

Page | 2

1.1.2 Gene mutation and cancer


Mutations are abnormal changes in the DNA of a gene. The building blocks of DNA are
called bases. The sequence of the bases determines the gene and its function. Mutations
involve changes in the arrangement of the bases that make up a gene. Even a change in just
one base among the thousands of bases that make up a gene can have a major effect.
Cells become cancer cells largely because of mutations in their genes. Often many mutations
are needed before a cell becomes a cancer cell. The mutations may affect different genes that
control cell growth and division. Some of these genes are called tumour suppressor genes.
Mutations may also cause some normal genes to become cancer-causing genes known as
oncogenes
Experts agree that it takes more than one mutation in a cell for cancer to occur. When
someone has inherited an abnormal copy of a gene, though, their cells already start out with
one mutation. This makes it all the easier (and quicker) for enough mutations to build up for
a cell to become cancer. That is why cancers that are inherited tend to occur earlier in life
than cancers of the same type that are not inherited. Even if you were born with healthy
genes, some of them can become changed (mutated) over the course of your life. These
acquired mutations and cause most cases of cancer. Some acquired mutations can be caused
by things that we are exposed to in our environment, including cigarette smoke, radiation,
hormones, and diet. Other mutations have no clear cause, and seem to occur randomly as the
cells divide. In order for a cell to divide to make 2 new cells, it has to copy its entire DNA.
With so much DNA, sometimes mistakes are made in the new copy. This leads to DNA
changes (mutations). Every time a cell divides, it is another opportunity for mutations to
occur. The numbers of gene mutations build up over time, which is why we have a higher
risk of cancer as we get older. It is important to realize that gene mutations happen in our
cells all the time. Usually, the cell detects the change and repairs it. If it cant be repaired, the
cell will get a signal telling it to die in a process called apoptosis. But if the cell doesn't die
and the mutation is not repaired, it may lead to a person developing cancer. This is more
likely if the mutation affects a gene involved with cell division or a gene that normally causes

Page | 3

a defective cell to die. Some people have a high risk of developing cancer because they have
inherited mutations in certain genes.

1.2 Oncogenes
An oncogene is a gene that has the potential to cause cancer. In tumour cells, they are often
mutated or expressed at high levels.
Most normal cells will undergo a programmed form of rapid cell death (apoptosis) when
critical functions are altered. Activated oncogenes can cause those cells designated for
apoptosis to survive and proliferate instead. Most oncogenes require an additional step, such
as mutations in another gene, or environmental factors, such as viral infection, to cause
cancer. Since the 1970s, dozens of oncogenes have been identified in human cancer. Many
cancer drugs target the proteins encoded by oncogenes.

1.3 Proto-oncogenes
A proto-oncogene is a normal gene that could become an oncogene due to mutations or
increased expression. The resultant protein encoded by an oncogene is termed oncoprotein.
Proto-oncogenes code for proteins that help to regulate cell growth and differentiation. Protooncogenes are often involved in signal transduction and execution of mitogenic signals,
usually through their protein products. Upon activation, a proto-oncogene (or its product)
becomes a tumour-inducing agent, an oncogene.

2. Historical Resume
The first theory of oncogenes was given by Danish physicist Niels Henrik Arley (1938-40)
scientific private secretary for Niels Bohr. 1947-53 head of Copenhagen University
geophysics research) at around 1950, but was rejected by contemporaries as nonsense. Later
on the term "oncogene" was rediscovered in 1969 by National Cancer Institute scientists
George Todaro and Robert Heubner.
The first confirmed oncogene was discovered in 1970 and was termed src (pronounced sarc
as in sarcoma). Src was in fact first discovered as an oncogene in a chicken retrovirus.

Page | 4

Experiments performed by Dr. G. Steve Martin of the University of California, Berkeley


demonstrated that the Src was indeed the oncogene of the virus. The first nucleotide sequence
of v-src was sequenced in 1980 by A.P. Czernilofsky et al.
In 1976 Drs. Dominique Stehelin, J. Michael Bishop and Harold E. Varmus of the University
of California, San Francisco demonstrated that oncogenes were activated proto-oncogenes,
found in many organisms including humans. For this discovery, proving Todaro and
Heubner's "oncogene theory", Bishop and Varmus were awarded the Nobel Prize in
Physiology or Medicine in 1989.
Oncoproteins are any proteins coded by an oncogene and they play an important role in the
regulation or synthesis of proteins linked to tumourigenic cell growth. Some oncoproteins are
accepted and used as tumour markers.

3. Classification
There are several systems for classifying oncogenes, but there is not yet a widely accepted
standard. They are sometimes grouped both spatially (moving from outside the cell inwards)
and chronologically (paralleling the "normal" process of signal transduction). There are
several categories that are commonly used:
Category

Examples

Cancers

Gene functions

Growth factors,
or mitogens

c-Sis

Glioblastomas,
fibrosarcomas,
osteosarcomas, breast
carcinomas, and
melanomas.

induces cell
proliferation.

Receptor
tyrosine kinases

epidermal growth factor


receptor (EGFR),
platelet-derived growth
factor receptor
(PDGFR), and vascular
endothelial growth
factor receptor
(VEGFR), HER2/neu

Breast cancer,
gastrointestinal stromal
tumours, non-small-cell
lung cancer and
pancreatic cancer

transduce signals
for cell growth and
differentiation.

Page | 5

Cytoplasmic
tyrosine kinases

Cytoplasmic
Serine/threonine
kinases and their
regulatory
subunits

Regulatory
GTPases

Transcription
factors

colorectal and breast


cancers, melanomas,
ovarian cancers, gastric
cancers, head and neck
cancers, pancreatic
cancer, lung cancer,
brain cancers, and blood
cancers

mediate the
responses to, and
the activation
receptors of cell
proliferation,
migration,
differentiation, and
survival

Raf kinase and cyclindependent kinases


(through
overexpression).

malignant melanoma,
papillary thyroid cancer,
colorectal cancer, and
ovarian cancer

Involved in
organism
development, cell
cycle regulation,
cell proliferation,
differentiation,
cells survival, and
apoptosis

Ras protein

adenocarcinomas of the
pancreas and colon,
thyroid tumours, and
myeloid leukemia

involved in
signaling a major
pathway leading to
cell proliferation.

myc gene

malignant T-cell
lymphomas and acute
myleoidleukemias,
breast cancer, pancreatic
cancer, retinoblastoma,
and small cell lung
cancer

-They regulate
transcription of
genes that induce
cell proliferation.

Src-family, Syk-ZAP-70
family, and BTK family
of tyrosine kinases, the
Abl gene in CML Philadelphia
chromosome

More detailed information for the above Table:

Growth factors are usually secreted by either specialized or not specialized cells to
induce cell proliferation in themselves, nearby cells, or distant cells. An oncogene
may cause a cell to secrete growth factors even though it does not normally do so. It
will thereby induce its own uncontrolled proliferation (autocrine loop), and
proliferation of neighboring cells. It may also cause production of growth hormones
in other parts of the body.

Page | 6

Receptor tyrosine kinases add phosphate groups to other proteins to turn them on or
off. Receptor kinases add phosphate groups to receptor proteins at the surface of the
cell (which receive protein signals from outside the cell and transmit them to the
inside of the cell). Tyrosine kinases add phosphate groups to the amino acid tyrosine
in the target protein. They can cause cancer by turning the receptor permanently on
(constitutively), even without signals from outside the cell.

Ras is a small GTPase that hydrolyses GTP into GDP and phosphate. Ras is activated
by growth factor signaling (i.e., EGF, TGFbeta) and acting like a binary switch
(on/off) in growth signaling pathways. Downstream effectors of Ras include three
mitogen-activated protein kinases Raf a MAP Kinase Kinase Kinase (MAPKKK),
MEK a MAP Kinase Kinase (MAPKK), and ERK a MAP Kinase (MAPK), which in
turn regulate genes that mediate cell proliferation.

4. Proto-oncogenes: A detailed account


Definitions:-There are trillions of living cells in the body that grow, divide, and die in an
orderly fashion. This process is tightly regulated by the genes within a cells nucleus. These
genes code for proteins that help regulate cell growth. These important genes are called
proto-oncogenes.
OR
A normally present gene that appears to have a role in the regulation of normal cell growth,
but that is converted to an oncogene by mutation.
A change in the DNA sequence of the proto-oncogene gives rise to an oncogene, which
produces a different protein and interferes with normal cell regulation.Proto-oncogenes have
many functions in a cell but they often code for proteins that stimulate cell division, prevent
cell differentiation or regulate programmed cell death (apoptosis). These are all essential
processes required for normal growth, development and the maintenance of healthy organs
and tissues. However, a mutated or defective version of a proto-oncogene (oncogene)
increases the production of these proteins, thereby leading to unregulated cell division, a

Page | 7

slower rate of cell differentiation and increased inhibition of cell death. Together, these
features define cells that have become cancerous.
Currently, about 40 genes are known to be proto-oncogenes. In 16 of them direct correlation
with tumour proliferation was shown, such as:

ERBB2 (HER2): breast cancer, by amplification;

KRAS: tumours of the esophagus, colon, pancreas, by point mutation;

beta-Catenin: Pancreatic cancer;

Cyclin E: liver tumours;

BRAF: melanomas;

BCR-ABL: chronic myeloid leukemia.

One example of a well known proto-oncogene is the HER2 gene. This gene codes for a
transmembrane tyrosine kinase receptor called human epidermal growth factor receptor
2.This protein receptor is involved in the growth, repair and division of cells in the breast. In
a healthy breast cell, there are two copies of HER2, but in some types of breast cancer, the
cells contain more than 2 copies, which lead to an excess production of the HER2 protein.
This causes the breast cells to grow, divide and proliferate much more quickly than healthy
breast cells.
This genetic defect in the breast cells is not inherited but is most likely to arise as a result of
aging. Researchers are still investigating whether environmental factors such as smoke and
pollution increase the likelihood of the defect occurring.
If a woman is diagnosed as HER2 positive, she has an abnormal number of HER2 genes and
is producing too much of the HER2 protein. This means cells in her breast are growing in an
upregulated manner and forming a cancerous tumour. If a woman is diagnosed as HER2
negative, then it is not HER2 protein production that is causing the cancer.

Page | 8

Another example of a proto-oncogene is the Myc gene, which codes for transcription factors.
When the gene sequence of Myc is altered, these transcription factors are produced at
increased rates and gene expression is altered giving rise to the formation of a tumour.

4.1 Activation of Proto-oncogenes to Oncogenes


The activation of oncogenes involves genetic changes to cellular proto-oncogenes. The
consequence of these genetic alterations is to confer a growth advantage to the cell. Three
genetic mechanisms activate oncogenes in human neoplasms:
1. Mutation
2. Gene amplification
3. Chromosome rearrangements.
These mechanisms result in either an alteration of proto-oncogene structure or an increase in
proto-oncogene expression. Because neoplasia is a multistep process, more than one of these
mechanisms often contributes to the genesis of human tumors by altering a number of
cancer-associated genes. Full expression of the neoplastic phenotype, including the capacity
for metastasis, usually involves a combination of proto-oncogene activation and tumor
suppressor gene loss or inactivation.

Figure: Schematic representation of the main mechanisms of oncogene activation (from


protooncogenes to oncogenes).

Page | 9

4.1.1 Mutation
Mutations activate protooncogenes through structural alterations in their encoded proteins.
These alterations, which usually involve critical protein regulatory regions, often lead to the
uncontrolled, continuous activity of the mutated protein. Various types of mutations, such as
base substitutions, deletions, and insertions, are capable of activating proto-oncogenes.
Retroviral oncogenes, for example, often have deletions that contribute to their activation.
Examples include deletions in the aminoterminal ligand-binding domains of the erb B, kit,
ros, met, and trk oncogenes. In human tumors, however, most characterized oncogene
mutations are base substitutions (point mutations) that change a single amino acid within the
protein.
Point mutations are frequently detected in the ras family of protooncogenes (K-ras, H-ras,
and N-ras). It has been estimated that as many as 15% to 20% of unselected human tumors
may contain a ras mutation. Mutations in K-ras predominate in carcinomas. Studies have
found K-ras mutations in about 30% of lung adenocarcinomas, 50% of colon carcinomas,
and 90% of carcinomas of the pancreas. N-ras mutations are preferentially found in
hematologic malignancies, with up to a 25% incidence in acute myeloid leukemias and
myelodysplastic syndromes. The majority of thyroid carcinomas have been found to have ras
mutations distributed among K-ras, H-ras, and N-ras, without preference for a single ras
family member but showing an association with the follicular type of differentiated thyroid
carcinomas. The majority of ras mutations involve codon 12 of the gene, with a smaller
number involving other regions such as codons 13 or 61. Ras mutations in human tumors
have been linked to carcinogen exposure. The consequence of ras mutations is the
constitutive activation of the signal-transducing function of the ras protein.
Another significant example of activating point mutations is represented by those affecting
the ret protooncogene in multiple endocrine neoplasia type 2A syndrome (MEN2A).

4.1.2 Activation by Amplification of Gene


Gene amplification refers to the expansion in copy number of a gene within the genome of a
cell. Gene amplification was first discovered as a mechanism by which some tumor cell lines

P a g e | 10

can acquire resistance to growth-inhibiting drugs. The process of gene amplification occurs
through redundant replication of genomic DNA, often giving rise to karyotypic abnormalities
called double-minute chromosomes (DMs) and homogeneous staining regions (HSRs). DMs
are characteristic minichromosome structures without centromeres. HSRs are segments of
chromosomes that lack the normal alternating pattern of light and dark-staining bands. Both
DMs and HSRs represent large regions of amplified genomic DNA containing up to several
hundred copies of a gene. Amplification leads to the increased expression of genes, which in
turn can confer a selective advantage for cell growth.
The frequent observation of DMs and HSRs in human tumors suggested that the
amplification of specific protooncogenes may be a common occurrence in neoplasia. Studies
then demonstrated that three proto-oncogene familiesmyc, erb B, and rasare amplified in a
significant number of human tumors. About 20% to 30% of breast and ovarian cancers show
c-myc amplification, and an approximately equal frequency of c-myc amplification is found
in some types of squamous cell carcinomas. N-myc was discovered as a new member of the
myc protooncogene family through its amplification in neuroblastomas. Amplification of Nmyc correlates strongly with advanced tumor stage in neuroblastoma, suggesting a role for
this gene in tumor progression. L-myc was discovered through its amplification in small-cell
carcinoma of the lung, a neuroendocrine-derived tumor. Amplification of erb B, the
epidermal growth factor receptor, is found in up to 50% of glioblastomas and in 10% to 20%
of squamous carcinomas of the head and neck. Approximately 15% to 30% of breast and
ovarian cancers have amplification of the erbB-2 (HER-2/neu) gene. In breast cancer, erbB-2
amplification correlates with advanced stage and poor prognosis. Members of the ras gene
family, including K-ras and N-ras, are sporadically amplified in various carcinomas.

4.1.3 Chromosomal Translocation


Chromosome translocations activate proto-oncogenes by one of two mechanisms
1. By generating an aberrant fusion gene comprised protein portions of two genes, usually
leading to constitutive activation of a kinase that was previously regulated by a signal. (BcrAbl)

P a g e | 11

2. By transcriptional deregulation of a proto-oncogene at one chromosome breakpoint by


juxtaposition with regulatory sequences from the other breakpoint. Thus, one gene and its
protein are normal; it is either over-expressed or expressed inappropriately leading to trouble.
(Myc)
Disease initiation by gene fusion: Philadelphia chromosome fusion protein of BCR-ABL
results in activation of proto-oncogene ABL
The normal ABL protein

a non-receptor protein tyrosine kinase

phosphorylation of ABL substrates promotes cell growth

enzymatic activity of normal ABL is tightly controlled

The BCR-ABL fusion protein

the tyrosine kinase activity cannot be downregulated due to replacement of N-terminal


ABL sequences with BCR sequences

the fusion protein constitutively promotes cell growth.

Thus BCR-ABL is the malignantly activated form of the ABL proto-oncogene.

Figure: The chromosome rearrangement in chronic myelogenous leukemia. The Philadelphia


chromosome, which is a diagnostic of CML, is a translocation between chromosomes 9 and 22.

P a g e | 12

Disease initiation by transcriptional deregulation: Burkitts lymphoma and MYC


Burkitts lymphoma is a B cell malignancy endemic in the malarial belt and common in
immunosuppressed populations.
Virtually all patients associated with BL have a common chromosomal breakpoint at t(8;14)
(q24;q32) which leads to the transcription of the Myc gene on chromosome 8 being
transcribed by an Immunoglobulin promoter on chromosome 14.

Figure: C-myc translocations found in Burkitt lymphoma. A, t(8;14)(q24;q32) translocation


involving the locus of immunoglobulin heavy-chain gene located at 14q32. B, t(8;14)(q24;q32)
translocation where only 2 exons (Ex) of c-myc are translocated under regulatory elements from
the immunoglobulin heavy-chain locus located at 14q32. C, t(8;22)(q24;q11) translocation
involving the l locus of immunoglobulin light-chain gene at 22q11. D, t(2;8)(p12;q24) translocation
involving the locus of immunoglobulin light chain gene located at 2p12.

5. Oncogenes: A detailed account


Definitions:-A gene that is a mutated (changed) form of a gene involved in normal cell
growth. Oncogenes may cause the growth of cancer cells. Mutations in genes that become
oncogenes can be inherited or caused by being exposed to substances in the environment that
cause cancer.

P a g e | 13

OR
A gene that played a normal role in the cell as a proto-oncogene and that has been altered by
mutation and now may contribute to the growth of a tumour.
OR
A gene that contributes to the production of cancer. Oncogenes are generally mutated forms
of normal cellular genes (proto-oncogenes). A gene capable, when activated, of transforming
a cell. Oncogenes are found in the oncogenically activated state in retroviruses and
transformed cells and in their normal non-oncogenically activated state in non-transformed
cells in which they are called proto-oncogenes.
OR
Oncogenes are the activated forms of normal cellular genes whose protein products are
involved in key signaling pathways governing cell survival, proliferation and differentiation.
Through a variety of mechanisms, including viral infection and chemical induced mutation,
these proteins become hyperactive or overexpressed and contribute to the cell growth and
behavior typical of cancer cells.
As study of the molecular and cellular basis of cancer progresses, it is becoming clear that no
two cancers are identical: tumour development is a complex process, and there are many
paths to malignancy. Nevertheless, certain tenets persist: that cancer arises as the result of
genetic change; that this leads to loss of control over cellular proliferation, and that usually
several genetic errors are required to reach the full neoplastic phenotype. Deregulated
cellular proliferation may arise in two main ways: through the loss of genes that normally
checkcell growth (the tumour suppressors) or by the gain of function of genes that either
promote cellular proliferation or prevent cell death (the oncogenes) (from Greek onkos,
tumour). Some cancer-associated genes, for example those involved in DNA repair, do not
fall easily into either category; others such as P53 are tumour suppressors in wild-type form,
but can act as dominant oncogenes in certain mutant forms. In many instances of adult
human cancer, both features oncogenic activation and loss of tumour suppressor activity
may be identified.

P a g e | 14

A typical oncogene has dominant activity, and requires only one allele to be activated in
order to be oncogenic. An oncogene may be viral or cellular in origin; a viral oncogene may
be unique to the virus, or a homologue of a cellular proto oncogene. An oncogene can
promote transformation in vitro, and tumour formation in transgenic animals; further, its
deregulation is usually recurrently associated with malignancy. Of over a hundred oncogenes
that have been described, many are not entirely typical, but the model remains useful.

5.1 Classes of Oncogenes


Roughly 100 different oncogenes have been identified. Few examples are enlisted here:
Oncogene

Location

Nuclear Transcription Regulators


jun
Nucleus
fos
Nucleus
erbA
Nucleus
Intracellular signal transducers

Function
Transcription factor
Transcription factor
Member of Steroid receptor family

abl
raf

Cytoplasm
Cytoplasm

Protein tyrosine kinase


Protein serine kinase

gsp
ras
Mitogens
sis
Mitogen receptor
erbB
fms
Apoptosis inhibitor
bcl2

Cytoplasm
Cytoplasm

G-protein subunit
GTP/GDP-binding protein

Extracellular

Secreted growth factor

Transmembrane
Transmembrane

Receptor tyrosine kinase


Receptor tyrosine kinase

Cytoplasm

Upstream inhibitor of caspase cascade

P a g e | 15

5.2 Viral Oncogenes


A true viral oncogene demonstrates the ability to transform primary cells in vitro, although
often cooperation between two oncogenes is required for efficient transformation. It should
cause tumour formation in transgenic mice, and be associated with cancers, either in its viral
form, or as deregulated expression of a cellular homologue.

5.2.1 Retroviral Oncogene


It seems that all oncogenes carried by retroviruses have a cellular counterpart, although in
many cases this has only been identified after discovery of the viral oncogene. Thus the RSV
(Rous Sarcoma virus) oncogene src (gene referring sarcoma) has a cellular homologue, SRC.
Despite the fact that RSV strains are not tumorigenic in mammals, mammalian fibroblasts
can be transformed to a malignant phenotype by both src and SRC; further, increased SRC
expression has been identified in some human cancers, including colon, skin and breast
cancers (Hesketh, 1997).

5.2.2 Viral Oncogenes in Humans


Epstein-Barr virus (EBV), Human Herpes virus 8 (HHV-8) and Human Pappilomavirus
(HPV) possess clearly defined oncogenes. EBV is a complex virus with a large genome; in
latent infections, about 11 viral genes are expressed, grouped into the nuclear antigens or
EBNAs (Epstein-Barr nuclear antigens), and the latent membrane proteins or LMPs. EBNA1
induces tumours in transgenic animals, but not transformation in vitro; EBNA2 has
transforming properties in vitro (Hesketh, 1997). It is likely that the oncogenic potential of
EBV is the result of the combined contribution of several of these protein products, some of
which activate cellular proliferative pathways, and others of which inhibit cell death. The
HHV-8 genome also contains a number of potential oncogenes: here, the protein products are
related to cyclin D, interleukins and interferon responsive factors. Two oncoproteins, E6 and
E7, are encoded within the genome of HPV types 16 and 18; these interfere with the function
of two important cellular tumour suppressors, p53 and Rb (zur Hausen, 1999). These
oncogenes result in tumour formation in transgenic mice, and cooperate with cellular genes
to transform fibroblasts in vitro. Other viral proteins contributing to human cancers are less

P a g e | 16

typical in their role as oncoproteins, or await clearer definition of their function in tumour
formation. HTLV-I (Human T-lymphoma virus-1) similarly possesses the viral gene, tax,
which transactivates the viral LTR (long terminal repeat) through proteinprotein interaction.
In addition it can transactivate a number of cellular genes, including those encoding
cytokines, cytokine receptors and transcription factors, and it is this function that is thought
to contribute to malignant transformation. The tax gene cannot, however, induce
transformation alone, and there may be other HTLV-I genes, and probably non-viral cellular
events, necessary for inducing the malignant T-cell phenotype. Similarly, HIV does not carry
a true oncogene, but it does carry the gene tat that encodes a protein with angiogenic
properties, inducing endothelial cell growth, migration and invasion in vitro.

5.3 Cellular Oncogenes


Viruses have contributed greatly to the present understanding of oncogene function, but have
had a less significant role in human oncogenesis. Most oncogenes involved in human
tumours are not viral but cellular genes that are present in the normal genome, which are
deregulated or activated by mutation. There are three mechanisms (discussed earlier) of
cellular oncogenic activation:
1. through alteration of the coding sequence by deletion or point mutation
2. by increasing the copy number of the gene(amplification)
3. by chromosomal rearrangement
Mechanism of Cellular Oncogene activation:
Malignancy

Oncogene

Mutation

Glioblastoma

EGFR

Amplification

Breast Carcinoma

ERB-B2

Amplification

Breast Carcinoma

CCND1

Amplification

Neuroblastoma

ERB-B2

Point Mutation

Pancreatic Carcinoma

RAS

Point Mutation

P a g e | 17

Coloretal Carcinoma

RAS

Point Mutation

Chronic Myeloid

BCR-ABL

Gene Fusion

Leukaemia
Burkitt
Lymphoma

MYC

IgH juxtaposition

Follicular Lymphoma

BCL2

IgH juxtaposition

Mantle Cell Lymphoma

CCND1

IgH juxtaposition

5.4 Normal functions of Oncogenes


Both cellular oncogenes and most viral oncogenes derive from normal cellular genes or
proto-oncogenes. These genes encode proteins that function in essential cellular pathways: in
general, a proto-oncogene is likely to be proliferative, and/or pro-survival, and/or an inhibitor
of differentiation. These functions of a cell are governed by factors in the extracellular
environment such as cellcell contact and growth factors, and signals are relayed from the
cell surface to the cell nucleus by signaling cascades.
Proto-oncogenes may be divided into broad groups according to their position in such
cascades: they may be growth factors; cell surface receptors; membrane transducers;
intracellular signalling proteins or transcription factors. The pathways are complex,
converging and diverging, and the endpoints proliferation, survival and state of
differentiation are often interdependent. Nevertheless, even an incomplete understanding of
normal proto-oncogene function provides insights into the role of oncogenes in
tumorigenesis.

5.4.1 Growth factors and Receptors


Cell surface receptors that function in growth regulation fall into several superfamilies. One
of these includes the platelet-derived growth factor (PDGF) receptor, the ligand for which
has an oncogenic homologue, SIS, which when overexpressed causes excessive signalling
through the PDGF pathway. Another of the receptor tyrosine kinase families is the epidermal
growth factor (EGF) receptor family. In this case, the gene encoding the EGF receptor itself

P a g e | 18

is amplified in some human tumours (Hanahan and Weinberg, 2000), causing it to function as
an oncogene. A related gene, NEU, is also overexpressed in tumours and has a viral
homologue erbB, which encodes a truncated EGF receptor. In this form, the receptor is
constitutively active, and does not require ligand binding to function. The EGF receptor, like
most growth factor receptors, is a tyrosine kinase, signalling to downstream molecules
through membrane-associated binding and phosphorylation

5.4.2 Membrane Transducers


Two important categories of protein that transmit signals from receptors to cytoplasmic
molecules are the non-receptor tyrosine kinases such as the protein encoded by SRC, and
GTP-binding proteins or G-proteins, such as members of the Ras family. This family is
comprised of closely related genes H-RAS, K-RAS and N-RAS, encoding proteins of 21 kDa
(hence, p21ras). Each p21ras protein binds one molecule of guanosine triphosphate (GTP) or
guanosine diphosphate (GDP); phosphorylation of bound GDP converts p21ras from off
mode to on mode, mediating binding and activation of downstream regulators like Raf
andMEK. RAS becomes oncogenic usually as the result of point mutations, which prevent
the hydrolysis of p21ras-bound GTP, maintaining it permanently in the on position.

5.4.3 Cell cycle regulators


The proliferation state of the cell is determined by its position in, and passage through, the
cell cycle. Passage through the cycle is in turn governed by a number of proteins that take
their cues from signalling cascades, allowing the cell to move from one stage to the next.
Some of these proteins can function as oncogenes such as cyclin D1, encoded by CCND1,
which normally governs entry of the cell into S phase from G1. When overexpressed as a
result of gene amplification or immunoglobulin H (IgH) translocation, the increased cyclin
D1 promotes cycling and proliferation.

5.4.4 Inhibitors of Apoptosis


In parallel with the mitogenic pathways are the pathways leading to cell death, which must be
inhibited in order for malignant proliferation to proceed in exponential fashion. Inhibitors of

P a g e | 19

apoptosis can thus function as oncogenes: the paradigm here is Bcl2, an antiapoptotic
molecule bound to the mitochondrial membrane, serving to prevent caspase activation. Bcl2
is overexpressed in many lymphomas (Hesketh, 1997).

5.4.5 Transcription factors


The endpoints of mitogenic, cell cycle and apoptotic signalling usually lie within the nucleus,
where expression of proteins is altered at the level of transcription. Many pathways converge
on transcription factors that promote or inhibit transcription of important genes. Not
surprisingly, several of these transcription factors are themselves proto-oncogenes. MYC, for
example, encodes an important transcription factor, regulating expression of many genes and
being essential for cell cycle progression. Similarly, fos and jun proteins are activated by a
number of mitogenic pathways. These proteins heterodimerize to each other and to other
transcription factors, and thus may act as AND gateways to protein expression, allowing
transcription of important genes when several growth signals are present.

6. Tumor Suppressor Genes


A tumor suppressor gene, or anti-oncogene, is a gene that protects a cell from one step on the
path to cancer. When this gene mutates to cause a loss or reduction in its function, the cell
can progress to cancer, usually in combination with other genetic changes. The loss of these
genes may be even more important than proto-oncogene/oncogene activation for the
formation of many kinds of human cancer cells. Tumor suppressor genes can be grouped into
categories including caretaker genes, gatekeeper genes, and landscaper genes; the
classification schemes are evolving as medicine advances, learning from fields including
molecular biology, genetics, and epigenetics.

6.1 Two-Hit Hypothesis


Unlike oncogenes, tumor suppressor genes generally follow the "two-hit hypothesis," which
implies that both alleles that code for a particular protein must be affected before an effect is
manifested. This is because if only one allele for the gene is damaged, the second can still

P a g e | 20

produce the correct protein. In other words, mutant tumor suppressors' alleles are usually
recessive whereas mutant oncogene alleles are typically dominant.
The two-hit hypothesis was first proposed by A.G. Knudson for cases of retinoblastoma.
Knudson observed that the age of onset of retinoblastoma followed 2nd order kinetics,
implying that two independent genetic events were necessary. He recognized that this was
consistent with a recessive mutation involving a single gene, but requiring bi-allelic
mutation. Oncogene mutations, in contrast, generally involve a single allele because they are
gain-of-function mutations.
There are exceptions to the "two-hit" rule for tumor suppressors, such as certain mutations in
the p53 gene product. p53 mutations can function as a "dominant negative," meaning that a
mutated p53 protein can prevent the function of normal protein from the un-mutated allele.
Other tumor-suppressor genes that are exceptions to the "two-hit" rule are those that exhibit
haploinsufficiency, including PTCH in medulloblastoma and NF1 in neurofibroma. An
example of this is the p27Kip1 cell-cycle inhibitor, in which mutation of a single allele
causes increased carcinogen susceptibility.

6.2 Functions
Tumor-suppressor genes, or more precisely, the proteins for which they code, either have a
damping or repressive effect on the regulation of the cell cycle or promote apoptosis, and
sometimes do both. The functions of tumor-suppressor proteins fall into several categories
including the following:
1. Repression of genes that are essential for the continuing of the cell cycle. If these genes
are not expressed, the cell cycle does not continue, effectively inhibiting cell division.
2. Coupling the cell cycle to DNA damage. As long as there is damaged DNA in the cell, it
should not divide. If the damage can be repaired, the cell cycle can continue.
3. If the damage cannot be repaired, the cell should initiate apoptosis (programmed cell
death) to remove the threat it poses for the greater good of the organisms produced

P a g e | 21

4. Some proteins involved in cell adhesion prevent tumor cells from dispersing, block loss
of contact inhibition, and inhibit metastasis. These proteins are known as metastasis
suppressors.
5. DNA repair proteins are usually classified as tumor suppressors as well, as mutations in
their genes increase the risk of cancer, for example mutations in HNPCC, MEN1 and
BRCA. Furthermore, increased mutation rate from decreased DNA repair leads to
increased inactivation of other tumor suppressors and activation of oncogenes.

6.3 Examples
The first tumor-suppressor protein discovered was the Retinoblastoma protein (pRb) in
human retinoblastoma; however, recent evidence has also implicated pRb as a tumor-survival
factor.
Another important tumor suppressor is the p53 tumor-suppressor protein encoded by the
TP53 gene. Homozygous loss of p53 is found in 65% of colon cancers, 3050% of breast
cancers, and 50% of lung cancers. Mutated p53 is also involved in the pathophysiology of
leukemias, lymphomas, sarcomas, and neurogenic tumors. Abnormalities of the p53 gene can
be inherited in Li-Fraumeni syndrome (LFS), which increases the risk of developing various
types of cancers.

6.4 p53
Tumor protein p53, also known as p53, cellular tumor antigen p53, phosphoprotein p53,
tumor suppressor p53, antigen NY-CO-13, or transformation-related protein 53 (TRP53), is
any isoform of a protein encoded by homologous genes in various organisms, such as TP53
(humans) and Trp53 (mice). This homolog (originally thought to be, and often spoken of as, a
single protein) is crucial in multicellular organisms, where it prevents cancer formation, thus,
functions as a tumor suppressor. As such, p53 has been described as "the guardian of the
genome" because of its role in conserving stability by preventing genome mutation. Hence
TP53 is classified as a tumor suppressor gene. (Italics are used to denote the TP53 gene name
and distinguish it from the protein it encodes.)

P a g e | 22

All these p53 proteins are called the p53 isoforms. The TP53 gene is the most frequently
mutated gene (>50%) in human cancer, indicating that the TP53 gene plays a crucial role in
preventing cancer formation. TP53 gene encodes proteins that bind to DNA and regulate
gene expression to prevent mutations of the genome.

6.4.1 Functions
p53 has many mechanisms of anticancer function and plays a role in apoptosis, genomic
stability, and inhibition of angiogenesis. In its anti-cancer role, p53 works through several
mechanisms:
1. It can activate DNA repair proteins when DNA has sustained damage. Thus, it may be an
important factor in aging.
2. It can arrest growth by holding the cell cycle at the G1/S regulation point on DNA
damage recognition (if it holds the cell here for long enough, the DNA repair proteins
will have time to fix the damage and the cell will be allowed to continue the cell cycle).
3. It can initiate apoptosis (i.e., programmed cell death) if DNA damage proves to be
irreparable.

P a g e | 23

Você também pode gostar