Mitogen

Last updated

A mitogen is a small bioactive protein or peptide that induces a cell to begin cell division, or enhances the rate of division (mitosis). Mitogenesis is the induction (triggering) of mitosis, typically via a mitogen. The mechanism of action of a mitogen is that it triggers signal transduction pathways involving mitogen-activated protein kinase (MAPK), leading to mitosis.

Contents

The cell cycle

Mitogens act primarily by influencing a set of proteins which are involved in the restriction of progression through the cell cycle. The G1 checkpoint is controlled most directly by mitogens: further cell cycle progression does not need mitogens to continue. The point where mitogens are no longer needed to move the cell cycle forward is called the "restriction point" and depends on cyclins to be passed. [1] One of the most important of these is TP53, a gene which produces a family of proteins known as p53. It, combined with the Ras pathway, downregulate cyclin D1, a cyclin-dependent kinase, if they are not stimulated by the presence of mitogens. In the presence of mitogens, sufficient cyclin D1 can be produced. This process cascades onwards, producing other cyclins which stimulate the cell sufficiently to allow cell division. While animals produce internal signals that can drive the cell cycle forward, external mitogens can cause it to progress without these signals. [2]

Endogenous mitogens

Mitogens can be either endogenous or exogenous factors. Endogenous mitogens function to control cell division is a normal and necessary part of the life cycle of multicellular organisms. For example, in zebrafish, an endogenous mitogen Nrg1 is produced in response to indications of heart damage. When it is expressed, it causes the outer layers of the heart to respond by increasing division rates and producing new layers of heart muscle cells to replace the damaged ones. This pathway can potentially be deleterious, however: expressing Nrg1 in the absence of heart damage causes uncontrolled growth of heart cells, creating an enlarged heart. [3] Some growth factors, such as vascular endothelial growth factor, are also capable of directly acting as mitogens, causing growth by directly inducing cell replication. This is not true for all growth factors, as some growth factors instead appear to cause mitogenic effects like growth indirectly by triggering other mitogens to be released, as evidenced by their lack of mitogenic activity in vitro, which VEGF has. [4] Other well-known mitogenic growth factors include platelet derived growth factor (PDGF) and epidermal growth factor (EGF). [5]

Relationship to cancer

Mitogens are important in cancer research due to their effects on the cell cycle. Cancer is in part defined by a lack of, or failure of, control in the cell cycle. This is usually a combination of two abnormalities: first, cancer cells lose their dependence on mitogens. Second, cancer cells are resistant to anti-mitogens.

Independence from mitogens

Rather than requiring endogenous or external mitogens to continue the cell cycle, cancer cells are able to grow, survive, and replicate without mitogens. Cancer cells may lose their dependence on external mitogens by a variety of pathways.

First, cancer cells can produce their own mitogens, a term called autocrine stimulation. [5] This can result in a deadly positive feedback loop - tumor cells produce their own mitogens, which stimulate more tumor cells to replicate, which can then produce even more mitogens. For example, consider one of the earliest oncogenes to be identified, p28sis from the simian sarcoma virus, which causes tumorigenesis in the host animal. Scientists found that p28sis has a nearly identical amino acid sequence as human platelet-derived growth factor (PDGF). [6] Thus, tumors formed by the simian sarcoma virus are no longer dependent on the fluctuations of PDGF that control cell growth; instead, they can produce their own mitogens in the form of p28sis. With enough p28sis activity, the cells can proliferate without restriction, resulting in cancer.

Second, cancer cells can have mutated cell-surface receptors for mitogens. The protein kinase domain found on mitogenic receptors is often hyperactivated in cancer cells, remaining turned on even in the absence of external mitogens. Additionally, some cancers are associated with an overproduction of mitogenic receptors on the cell surface. With this mutation, cells are stimulated to divide by abnormally low levels of mitogens. One such example is HER2, a receptor tyrosine kinase that responds to the mitogen EGF. Overexpression of HER2 is common in 15-30% of breast cancers, [7] allowing the cell cycle to progress even with extremely low concentrations of EGF. The overexpression of kinase activity in these cells aids in their proliferation. These are known as hormone-dependent breast cancers, as the kinase activation in these cancers is connected to exposure to both growth factors and estradiol. [8]

Third, downstream effectors of mitogenic signaling are often mutated in cancer cells. An important mitogenic signaling pathway in humans is the Ras-Raf-MAPK pathway. Mitogenic signaling normally activates Ras, a GTPase, that then activates the rest of the MAPK pathway, ultimately expressing proteins that stimulate cell cycle progression. It is likely that most, if not all, cancers have some mutation in the Ras-Raf-MAPK pathway, most commonly in Ras. [5] These mutations allow the pathway to be constitutively activated, regardless of the presence of mitogens.

Resistance to anti-mitogens

Cell proliferation is often regulated by not only external mitogens but also by anti-mitogens, which inhibit cell cycle progression past G1. In normal cells, anti-mitogenic signaling as a result of DNA damage, preventing the cells from replicating and dividing. Tumor cells that are resistant to anti-mitogens allow the cell cycle to move forward when it should be prevented by some anti-mitogenic mechanism. This resistance to anti-mitogens might simply arise from overstimulation by positive mitogens. In other cases, tumor cells possess loss-of-function mutations in some part of the anti-mitogenic pathway. For example, consider the well-known anti-mitogen, transforming growth factor (TGF-𝝱). TGF-𝝱 works by binding to cell-surface receptors and activating the Smad gene regulatory proteins. Smad proteins then trigger an increase in p15, which inhibits cyclin D1 and prevents cell cycle progression. In many cancers, there is a loss-of-function mutation in the Smad proteins, thus negating the entire anti-mitogenic pathway. [5]

Multiple mutations required

Not just one but multiple mitogenic mutations are required for cancer to proliferate. Generally, multiple mutations in different subsystems (an oncogene and a tumor suppressor gene) are the most effective at causing cancer. For example, a mutation that hyperactivates the oncogene Ras and another that inactivates the tumor suppressor pRb is far more tumorigenic than either protein alone. [5] Tumor cells are also resistant to the hyperproliferation stress response. Normal cells have apoptotic proteins that will respond to an overstimulation of mitogenic signaling pathways by triggering cell death or senescence. This generally prevents the onset of cancer from a single oncogenic mutation. In tumor cells, there is generally another mutation that inhibits apoptotic proteins as well, suppressing the hyperproliferation stress response. [5]

Use in immunology

Lymphocytes can enter mitosis when they are activated by mitogens or antigens. B cells specifically can divide when they encounter an antigen matching their immunoglobulin. T cells undergo mitosis when stimulated by mitogens to produce small lymphocytes that are then responsible for the production of lymphokines, which are substances that modify the host organism to improve its immunity. B cells, on the other hand, divide to produce plasma cells when stimulated by mitogens, which then produce immunoglobulins, or antibodies. [9] Mitogens are often used to stimulate lymphocytes and thereby assess immune function. The most commonly used mitogens in clinical laboratory medicine are:

NameActs upon T cells?Acts upon B cells?
phytohaemagglutinin (PHA)yesno
concanavalin A (conA)yesno
lipopolysaccharide (LPS)noyes
pokeweed mitogen (PWM)yes [10] yes

Lipopolysaccharide toxin from gram-negative bacteria is thymus-independent. They may directly activate B cells, regardless of their antigenic specificity. Plasma cells are terminally differentiated and, therefore, cannot undergo mitosis. Memory B cells can proliferate to produce more memory cells or plasma B cells. This is how the mitogen works, that is, by inducing mitosis in memory B cells to cause them to divide, with some becoming plasma cells.[ citation needed ]

Other uses

Mitogen-activated protein kinase (MAPK) pathways can induce enzymes such as the COX-2 enzyme. [11] MAPK pathways may also play a role in the regulation of PTGS2. [12]

See also

Related Research Articles

Oncogene Gene that has the potential to cause cancer

An oncogene is a gene that has the potential to cause cancer. In tumor cells, these genes are often mutated, or expressed at high levels.

Kinase Enzyme catalyzing transfer of phosphate groups onto specific substrates

In biochemistry, a kinase is an enzyme that catalyzes the transfer of phosphate groups from high-energy, phosphate-donating molecules to specific substrates. This process is known as phosphorylation, where the high-energy ATP molecule donates a phosphate group to the substrate molecule. This transesterification produces a phosphorylated substrate and ADP. Conversely, it is referred to as dephosphorylation when the phosphorylated substrate donates a phosphate group and ADP gains a phosphate group. These two processes, phosphorylation and dephosphorylation, occur four times during glycolysis.

Paracrine signaling

Paracrine signaling is a form of cell signaling, a type of cellular communication in which a cell produces a signal to induce changes in nearby cells, altering the behaviour of those cells. Signaling molecules known as paracrine factors diffuse over a relatively short distance, as opposed to cell signaling by endocrine factors, hormones which travel considerably longer distances via the circulatory system; juxtacrine interactions; and autocrine signaling. Cells that produce paracrine factors secrete them into the immediate extracellular environment. Factors then travel to nearby cells in which the gradient of factor received determines the outcome. However, the exact distance that paracrine factors can travel is not certain.

A mitogen-activated protein kinase is a type of protein kinase that is specific to the amino acids serine and threonine. MAPKs are involved in directing cellular responses to a diverse array of stimuli, such as mitogens, osmotic stress, heat shock and proinflammatory cytokines. They regulate cell functions including proliferation, gene expression, differentiation, mitosis, cell survival, and apoptosis.

Restriction point Animal cell cycle checkpoint

The restriction point (R), also known as the Start or G1/S checkpoint, is a cell cycle checkpoint in the G1 phase of the animal cell cycle at which the cell becomes "committed" to the cell cycle, and after which extracellular signals are no longer required to stimulate proliferation. The defining biochemical feature of the restriction point is the activation of G1/S- and S-phase cyclin-CDK complexes, which in turn phosphorylate proteins that initiate DNA replication, centrosome duplication, and other early cell cycle events. It is one of three main cell cycle checkpoints, the other two being the G2-M DNA damage checkpoint and the spindle checkpoint.

Nerve growth factor Mammalian protein found in Homo sapiens

Nerve growth factor (NGF) is a neurotrophic factor and neuropeptide primarily involved in the regulation of growth, maintenance, proliferation, and survival of certain target neurons. It is perhaps the prototypical growth factor, in that it was one of the first to be described. Since it was first isolated by Nobel Laureates Rita Levi-Montalcini and Stanley Cohen in 1956, numerous biological processes involving NGF have been identified, two of them being the survival of pancreatic beta cells and the regulation of the immune system.

Biological crosstalk refers to instances in which one or more components of one signal transduction pathway affects another. This can be achieved through a number of ways with the most common form being crosstalk between proteins of signaling cascades. In these signal transduction pathways, there are often shared components that can interact with either pathway. A more complex instance of crosstalk can be observed with transmembrane crosstalk between the extracellular matrix (ECM) and the cytoskeleton.

Mitogen Activated Protein (MAP) kinase kinase kinase, MAPKKK is a serine/threonine-specific protein kinase which acts upon MAP kinase kinase. Subsequently, MAP kinase kinase activates MAP kinase. Several types of MAPKKK can exist but are mainly characterized by the MAP kinases they activate. MAPKKKs are stimulated by a large range of stimuli, primarily environmental and intracellular stressors. MAPKKK is responsible for various cell functions such as cell proliferation, cell differentiation, and apoptosis. The duration and intensity of signals determine which pathway ensues. Additionally, the use of protein scaffolds helps to place the MAPKKK in close proximity with its substrate to allow for a reaction. Lastly, because MAPKKK is involved in a series of several pathways, it has been used as a therapeutic target for cancer, amyloidosis, and neurodegenerative diseases. In humans, there are at least 19 genes which encode MAP kinase kinase kinases:

The MAPK/ERK pathway is a chain of proteins in the cell that communicates a signal from a receptor on the surface of the cell to the DNA in the nucleus of the cell.

Platelet-derived growth factor receptor

Platelet-derived growth factor receptors (PDGF-R) are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family. PDGF subunits -A and -B are important factors regulating cell proliferation, cellular differentiation, cell growth, development and many diseases including cancer. There are two forms of the PDGF-R, alpha and beta each encoded by a different gene. Depending on which growth factor is bound, PDGF-R homo- or heterodimerizes.

In molecular biology, extracellular signal-regulated kinases (ERKs) or classical MAP kinases are widely expressed protein kinase intracellular signalling molecules that are involved in functions including the regulation of meiosis, mitosis, and postmitotic functions in differentiated cells. Many different stimuli, including growth factors, cytokines, virus infection, ligands for heterotrimeric G protein-coupled receptors, transforming agents, and carcinogens, activate the ERK pathway.

Cyclin D

Cyclin D is a member of the cyclin protein family that is involved in regulating cell cycle progression. The synthesis of cyclin D is initiated during G1 and drives the G1/S phase transition. Cyclin D protein is anywhere from 155 to 477 amino acids in length.

The ErbB family of proteins contains four receptor tyrosine kinases, structurally related to the epidermal growth factor receptor (EGFR), its first discovered member. In humans, the family includes Her1, Her2, Her3 (ErbB3), and Her4 (ErbB4). The gene symbol, ErbB, is derived from the name of a viral oncogene to which these receptors are homologous: erythroblastic leukemia viral oncogene. Insufficient ErbB signaling in humans is associated with the development of neurodegenerative diseases, such as multiple sclerosis and Alzheimer's disease, while excessive ErbB signaling is associated with the development of a wide variety of types of solid tumor.

ELK1 Protein-coding gene in the species Homo sapiens

ETS Like-1 protein Elk-1 is a protein that in humans is encoded by the ELK1. Elk-1 functions as a transcription activator. It is classified as a ternary complex factor (TCF), a subclass of the ETS family, which is characterized by a common protein domain that regulates DNA binding to target sequences. Elk1 plays important roles in various contexts, including long-term memory formation, drug addiction, Alzheimer's disease, Down syndrome, breast cancer, and depression.

MAPK7

Mitogen-activated protein kinase 7 also known as MAP kinase 7 is an enzyme that in humans is encoded by the MAPK7 gene.

Trk receptors are a family of tyrosine kinases that regulates synaptic strength and plasticity in the mammalian nervous system. Trk receptors affect neuronal survival and differentiation through several signaling cascades. However, the activation of these receptors also has significant effects on functional properties of neurons.

MAP2K5

Dual specificity mitogen-activated protein kinase kinase 5 is an enzyme that in humans is encoded by the MAP2K5 gene.

MAPK4

Mitogen-activated protein kinase 4 is an enzyme that in humans is encoded by the MAPK4 gene.

The Akt signaling pathway or PI3K-Akt signaling pathway is a signal transduction pathway that promotes survival and growth in response to extracellular signals. Key proteins involved are PI3K and Akt.

Tyrosine phosphorylation Phosphorylation of peptidyl-tyrosine

Tyrosine phosphorylation is the addition of a phosphate (PO43−) group to the amino acid tyrosine on a protein. It is one of the main types of protein phosphorylation. This transfer is made possible through enzymes called tyrosine kinases. Tyrosine phosphorylation is a key step in signal transduction and the regulation of enzymatic activity.

References

  1. Bohmer et al. "Cytoskeletal Integrity Is Required throughout the Mitogen Stimulation Phase of the Cell Cycle and Mediates the Anchorage-dependent Expression of Cyclin DI". January 1996, Molecular Biology of the Cell, Vol. 7, pp. 101-111.
  2. Foijer et al. "Mitogen requirement for cell cycle progression in the absence of pocket protein activity". December 2005, Cancer Cell, Vol. 8, pp. 455-466
  3. Gemberling et al. "Nrg1 is an injury-induced cardiomyocyte mitogen for the endogenous heart regeneration program in zebrafish". 1 April 2015, eLifeSciences.
  4. Leung et al. "Vascular Endothelial Growth Factor is a Secreted Angiogenic Mitogen". 8 December 1989, Science, Vol. 246, pp 1306-1309.
  5. 1 2 3 4 5 6 Morgan, David (2007). “The Cell Cycle: Principles of Control”. New Science Press.
  6. Waterfield, M., Scrace, G., Whittle, N. et al. Platelet-derived growth factor is structurally related to the putative transforming protein p28sis of simian sarcoma virus. Nature 304, 35–39 (1983) doi:10.1038/304035a0
  7. Mitri Z, Constantine T, O'Regan R (2012). "The HER2 Receptor in Breast Cancer: Pathophysiology, Clinical Use, and New Advances in Therapy". Chemotherapy Research and Practice. 2012: 743193
  8. Santen et al. "The role of mitogen-activated protein (MAP) kinase in breast cancer". February 2002, The Journal of Steroid Biochemistry and Molecular Biology, Vol. 80, pp. 239-256
  9. Barret, James (1980). Basic Immunology and its Medical Application (2 ed.). St.Louis: The C.V. Mosby Company. pp. 52–3. ISBN   978-0-8016-0495-9.
  10. Assenmacher, Mario; Avraham, Hava Karsenty; Avraham, Shalom; Bala, Shukal, eds. (2005), "Pokeweed Mitogen", Encyclopedic Reference of Immunotoxicology, Springer, p. 509, doi:10.1007/3-540-27806-0_1183, ISBN   978-3-540-27806-1
  11. Font-Nieves, M; Sans-Fons, MG (2012). "Induction of COX-2 enzyme and down-regulation of COX-1 expression by lipopolysaccharide (LPS) control prostaglandin E2 production in astrocytes". Journal of Biological Chemistry. 287 (9): 6454–68. doi: 10.1074/jbc.M111.327874 . PMC   3307308 . PMID   22219191.
  12. Casciani, V; Marinoni, E (2008). "Opposite effect of phorbol ester PMA on PTGS2 and PGDH mRNA expression in human chorion trophoblast cells". Reproductive Sciences. 15 (1): 40–50. doi:10.1177/1933719107309647. PMID   18212353. S2CID   10706385.