Common Signaling Pathways Used During Development





During the process of embryonic development, undifferentiated precursor cells differentiate and organize into the complex structures found in functional adult tissues. This intricate process requires cells to integrate many intrinsic and extrinsic cues for development to occur properly. These cues control the proliferation, differentiation, and migration of cells to determine the final size and shape of the developing organs. Disruption of these signaling pathways can result in human developmental disorders and birth defects. These key developmental signaling pathways are also frequently co-opted in the adult by diseases such as cancer.


Given the diverse changes that occur during embryogenesis, it may appear that a correspondingly diverse set of signaling pathways should regulate these processes. In contrast, the differentiation of many cell types is regulated through a relatively restricted set of molecular signaling pathways:




  • Intercellular communication: Development involves the interaction of a cell with neighboring cell directly (gap junctions) or indirectly (cell adhesion molecules).



  • Morphogens: These diffusible molecules specify which cell type is generated at a specific anatomic location and direct the migration of cells and their processes to their final destinations. Morphogens include retinoic acid; the transforming growth factor-β (TGF-β) superfamily of proteins, including bone morphogenetic proteins (BMPs); and WNT protein families. Table 21.1 explains gene and protein nomenclature.



    Table 21.1

    International Nomenclature Standards for Genes and Proteins





















    Gene Human Italic, all letters capitalized PAX6
    Mouse Italic, first letter capitalized Pax6
    Protein Human Roman, all letters capitalized PAX6
    Mouse Roman, all letters capitalized PAX6



  • Hedgehog: The hedgehog family of morphogens signaling pathway in human cells is localized to a structure called the primary cilium. Disruption of the components of the hedgehog pathway result in a set of diseases termed ciliopathies.



  • Receptor tyrosine kinases (RTKs): Many growth factors signal by binding to and activating membrane-bound RTKs. These kinases are essential for the regulation of cellular proliferation, apoptosis, and migration, as well as processes such as the growth of new blood vessels and axonal processes in the nervous system.



  • Notch-Delta: This pathway often specifies which cell fate the precursor cells adopt.



  • Transcription factors: These evolutionarily conserved proteins activate or repress downstream genes that are essential for many cellular processes. Many transcription factors are members of the homeobox (HOX) or helix-loop-helix (HLH) families. Their activity can be regulated by all of the other pathways described in this chapter.



  • Epigenetic effects: These heritable changes in gene function do not result from a change in DNA sequence. Examples of epigenetic modifications are histone acetylation, histone methylation, microRNAs (miRNAs) and DNA methylation.



  • Stem cells: Embryonic stem cells can give rise to all cells and tissues in the developing organism. Adult stem cells maintain tissue homeostasis in the mature organism. These types of stem cells and induced pluripotent stem cells (iPS) are potential sources of cells for the regeneration and repair of injured or degenerating cells and organs. iPS derived from patient cells can be used to model developmental processes in vitro and to screen potential therapies. New advancements in gene editing have greatly enhanced our ability to model human diseases both in vitro and in vivo.



Intercellular Communication


During embryonic development, cells receive signals from their external environment and communicate with neighboring cells. This communication directs the cell to undergo processes such as proliferation, differentiation, and migration. Two classes of proteins required for intercellular communication are discussed: gap junctions and cell adhesion molecules.


Gap Junctions


Gap junctions are a means for cells to directly communicate with one another; this is known as gap junction intercellular communication (GJIC). Although the pore size of the channels vary, only small molecules (e.g., second messengers, ions such as calcium, adenosine triphosphate [ATP]) less than 1 kDa can pass through, which excludes most proteins and nucleic acids. In the nervous and cardiac systems, gap junctions help to establish electrical cell coupling (“electrical” synapse).


Although the function of gap junctions is quite straightforward, the structure of these intercellular channels is complex and highly regulated throughout development ( Fig. 21.1 ). Each gap junction is composed of two hemichannels known as connexons. Each hexameric connexon consists of six individual connexin subunits. An individual connexin (Cx) molecule consists of four transmembrane domains. Vertebrates have more than 20 connexin molecules. Cellular and tissue functional diversity of gap junctions depends on whether individual connexons are the same (homotypic) or different (heterotypic) and whether each connexon is made from the same (homomeric) or different (heteromeric) connexin molecules.




Fig. 21.1


Gap junction intercellular communication. A , The connexin molecule consists of four transmembrane domains and two extracellular domains, and its N- and C-termini are cytoplasmic. B , Connexons, or hemichannels, are hexameric structures consisting of six connexin subunits. A gap junction can be formed from two homophilic or heterophilic connexons. Small molecules (e.g., ions, adenosine triphosphate [ATP]) less than 1 kDa can pass through an open gap junction.


Early in development, GJIC is important for the rapid distribution of ions and other molecules essential for regionalization before the establishment of distinct boundaries and compartments. The importance of GJIC has been demonstrated in the developing chick hindbrain (rhombencephalon) by a combination of dye transfer and electrical coupling studies.


Some of the better-characterized connexins include Cx43 (heart, brain), Cx45 (heart, pancreas), Cx32 (myelin), and Cx36 (pancreas, brain). In this nomenclature system, the number following Cx refers to the molecular weight in kilodaltons (kDa) of the proteins. Mutations in Cx genes result in diseases such as the hereditary peripheral neuropathy X-linked Charcot–Marie–Tooth disease ( GJB1 , formerly CX32 ). It was previously thought that connexons had to bind to a connexon on an adjacent cell to functionally signal. However, it has since been shown that unbound connexons (hemichannels) enable the exchange of ions and small molecules between the cytoplasm and the extracellular space, especially during pathophysiologic conditions. Aberrant hemichannel activation through GJB2 (formerly CX26 ) can result in keratitis-ichthyosis-deafness syndrome.


Cell Adhesion Molecules


Cell adhesion molecules have large extracellular domains that interact with extracellular matrix (ECM) components or adhesion molecules on neighboring cells. These molecules often contain a transmembrane segment and a short cytoplasmic domain that regulate intracellular signaling cascades. Two classes of molecules that have important roles during embryonic development are cadherins and members of the immunoglobulin superfamily of cell adhesion molecules.


Cadherins


Cadherins are critical for embryonic morphogenesis because they regulate the separation of cell layers (endothelial and epidermal), cell migration, cell sorting, establishment of well-defined boundaries, synaptic connections, and the growth cones of neurons. Cadherins mediate the interaction between the cell and its extracellular milieu (neighboring cells and ECM).


Cadherins were originally classified by their site of expression. E-cadherin (epithelial cadherin) is highly expressed in epithelial cells, whereas N-cadherin (neural cadherin) is highly expressed in neural cells.


Cadherins mediate homophilic, calcium-dependent binding. A typical cadherin molecule has a large extracellular domain, a transmembrane domain, and an intracellular tail ( Fig. 21.2 ). The extracellular domain contains five extracellular repeats (EC repeats) and has four Ca 2+ -binding sites. Cadherins form dimers that interact with cadherin dimers in adjacent cells. These complexes are found clustered in adherens junctions , which result in the formation of a tight barrier between epithelial or endothelial cells.




Fig. 21.2


Structure of cadherin and neural cell adhesion molecule (NCAM). A , The cadherin extracellular domain contains four calcium-binding sites and five repeated domains called extracellular cadherin domains. Each cadherin molecule forms a homodimer. On the intracellular domain, cadherin binds directly to p120 catenin and to β-catenin, which binds to α-catenin. This complex links the cadherin molecules to the actin cytoskeleton. B , Extracellularly, NCAM contains five immunoglobulin repeats and two fibronectin type III domains. The fifth immunoglobulin repeat is modified by polysialylation, which decreases the adhesiveness of the NCAM molecule. Intracellular signaling is transmitted by the FYN and FAK kinases.


Through its intracellular domain, cadherins bind to p120 catenin, β-catenin, and α-catenin. These proteins connect cadherin to the cytoskeleton. E-cadherin expression is lost as epithelial cells transition to mesenchymal cells ( epithelial to mesenchymal transition [EMT] ). EMT is required for the formation of neural crest cells during development, and the same process can occur in tumors that develop from epithelial cell types.


Immunoglobulin Superfamily


There are more than 700 members of the immunoglobulin superfamily of cell adhesion molecules in the human genome. This large family of proteins is involved in a wide variety of cellular processes. One member of this class, the neural cell adhesion molecule (NCAM), is an abundant protein in the brain and has three isoforms that result from alternative splicing. It has a large extracellular domain that contains five immunoglobulin (Ig) repeats and two fibronectin domains (see Fig. 21.2 ). This region mediates the calcium-independent homophilic binding of NCAM to itself and heterophilic binding to other cell adhesion molecules (L1 and TAG1), RTKs (fibroblast growth factor receptor [FGFR]), or the ECM. Ligand binding induces intracellular signaling through the FYN and FAK intracellular kinases.


NCAM undergoes polysialylation (PSA), a unique post-translational glycosylation modification. PSA-NCAM is abundant early in neural development and becomes restricted to areas of neural plasticity and migration in the adult. It is thought that PSA decreases the adhesiveness of NCAM and facilitates migration. NCAM regulates neurite outgrowth, axonal pathfinding, survival, and plasticity.




Morphogens


Extrinsic signals guide the differentiation and migration of cells during development and thereby dictate the morphology and function of developing tissues (see Chapter 5 ). Many of these morphogens are found in concentration gradients in the embryo, and different morphogens can be expressed in opposing gradients in the dorsal-ventral, anterior-posterior, proximal-distal, and medial-lateral axes. The fate of a specific cell can be determined by its location along these gradients. Cells can be attracted or repelled by morphogens depending on the particular set of receptors expressed on the cell surface.


Retinoic Acid


The anterior (rostral, head)–posterior (caudal, tail), or anteroposterior, axis of the embryo is crucial for determining the correct location for structures such as limbs and for the patterning of the nervous system. For decades, it has been clinically evident that alterations in the level of vitamin A (retinol) in the diet (excessive or insufficient amounts) can lead to the development of congenital malformations (see Chapters 17 and 20 ).


The bioactive form of vitamin A is retinoic acid , which is formed by the oxidation of retinol to retinal by retinol dehydrogenases and the subsequent oxidation of retinal by retinal aldehyde dehydrogenase. Free levels of retinoic acid can be further modulated by cellular retinoic acid–binding proteins that sequester retinoic acid. Retinoic acid also can be actively degraded into inactive metabolites by enzymes such as CYP26 ( Fig. 21.3 ). Normally, retinoic acid posteriorizes the body plan. Excessive retinoic acid levels or inhibition of retinoic acid degradation both lead to a truncated body axis in which structures have a more posterior nature. Insufficient retinoic acid or defects in enzymes such as retinal aldehyde dehydrogenase lead to a more anteriorized structure.




Fig. 21.3


Regulation of retinoic acid metabolism and signaling. Dietary retinol (vitamin A) is converted to retinal by the action of retinol dehydrogenases. The concentration of free retinal is controlled by the action of cellular retinal-binding proteins. Similarly, retinal is converted to retinoic acid by retinal dehydrogenases, and its free level is modulated by sequestration by cellular retinoic acid–binding proteins and degradation by CYP26. The bioactive form of retinoic acid is all- trans retinoic acid.


At a molecular level, retinoic acid binds to its receptors inside the cell and activates them. Retinoic acid receptors are transcription factors, and their activation regulates the expression of downstream genes. Crucial targets of retinoic acid receptors in development are the HOX genes. Due to their profound influence on early development, retinoids are powerful teratogens, especially during the first trimester.


Transforming Growth Factor-β and Bone Morphogenetic Proteins


Members of the TGF-β superfamily include TGF-β, BMPs, activin, and nodal. These molecules contribute to the establishment of dorsoventral patterning, cell fate decisions, and formation of specific organs, including the nervous system, kidneys, skeleton, and blood (see Chapter 5, Chapter 16, Chapter 17 ).


In humans, there are three TGF-β isoforms (TGF-β 1 , TGF-β 2 , and TGF-β 3 ). Binding of these ligands to heterotetrameric (four-subunit) complexes, consisting of specific type I (inactive kinase domain) and type II TGF-β receptor subunit (TβR-II) (constitutively active) transmembrane serine-threonine kinase receptors results in intracellular signaling events ( Fig. 21.4 ). When TGF-β ligands bind to their respective membrane-bound type II receptor, a type I receptor is recruited and transphosphorylated, and its kinase domain is activated, subsequently phosphorylating intracellular receptor-associated SMAD proteins (R-SMADs).




Fig. 21.4


Transforming growth factor-β (TGF-β) /SMAD signaling pathway. A , The type II TGF-β receptor subunit (TβR-II) is constitutively active. B , On binding of ligand to TβR-II, a type I receptor subunit is recruited to form a heterodimeric receptor complex, and the TβR-I kinase domain is transphosphorylated (-P). Signaling from the activated receptor complex phosphorylates R-SMADs, which then bind to a co-SMAD, translocate from the cytoplasm to the nucleus, and activate gene transcription with cofactors (X).


The SMADs are a large family of intercellular proteins that are divided into three classes: receptor activated (R-SMADs 1–3, 5, and 8), common partner (co-SMADs such as SMAD4), and inhibitory SMADs (I-SMADs such as SMAD6 and SMAD7). R-SMAD/SMAD4 complexes translocate to the nucleus and regulate target gene transcription by interacting with other proteins or function as transcription factors by directly binding to DNA.


Inhibitory SMAD proteins block the actions of I-SMAD by several mechanisms, such as preventing phosphorylation of R-SMADs by TβR-I, induction of R-SMAD degradation, and transcriptional repression. TβR-I activation is a highly regulated process that involves membrane-anchored coreceptors and other receptor-like molecules that can sequester ligands and prevent their binding to respective TβR-II receptors. Dominant negative forms of TβR-II have inactive kinase domains and cannot transphosphorylate TβR-I, thereby blocking downstream signaling events. The diversity of TGF-β ligand, TβR-I and TβR-II, coreceptor, ligand trap, and R-SMAD combinations contributes to particular developmental and cell-specific processes, often in combination with other signaling pathways.


Hedgehog and the Primary Cilium


The sonic hedgehog gene (SHH) was the first identified mammalian ortholog of the Drosophila hedgehog gene (Hh) . SHH and other related proteins, including desert hedgehog and Indian hedgehog, are secreted morphogens critical to early patterning, cell migration, and differentiation of many cell types and organ systems (see Chapter 5 ).


In Drosophila , cells have various thresholds for response to the secreted Hh signal. The primary receptor for Shh is Patched (PTCH in humans, PTC family in mice), a 12-transmembrane domain protein that in the absence of Shh inhibits Smoothened (Smo), a seven-transmembrane domain G protein–linked protein, and downstream signaling to the nucleus. However, in the presence of Shh, Patched (Ptc) inhibition is blocked, and downstream events follow, including nuclear translocation of Gli (Gli1, Gli2, Gli3) with transcriptional activation of target genes such as Ptc1 , Engrailed , and others ( Fig. 21.5 ).




Fig. 21.5


Sonic hedgehog/Patched signaling pathway in vertebrates. A , The Patched (Ptc) receptor inhibits signaling from the Smoothened (Smo) receptor. In a complex with the kinesin-like protein Costal 2 (Cos2) and serine-threonine kinase Fused (Fu), Gli is modified to become a transcriptional repressor (Gli-R). B , Sonic hedgehog (Shh) is cleaved, and cholesterol (Chol) is added to its N-terminus. This modified Shh ligand inhibits the Ptc receptor, permitting Smo signaling, and activated Gli (Gli-A) ultimately translocates to the nucleus to activate target genes with cyclic AMP–binding protein (CBP). In vertebrates, SHH signaling takes place in primary cilia (inset). CK1 , Casein kinase 1; GSK3 , glycogen synthase kinase 3; P , phosphate group; PKA , protein kinase A; SuFu , suppressor of Fused.


Other membrane-bound SHH coreceptors have been identified with key roles in ventral neural patterning, including BOC, GAS1, and the LDL receptor-related protein 2 (LRP2; in mammals). Individually, these coreceptors act to enhance SHH signaling. BOC and GAS1 each interact with the SHH canonical receptor PTC/PTCH to form distinct receptor complexes essential for cell proliferation mediated by SHH. The role of BOC is especially important for commissural axonal guidance during development and in medulloblastoma progression. In contrast, LRP2 promotes the internalization and subsequent degradation of PTC/PTCH upon SHH binding thereby removing PTC/PTCH inhibition of Smo. Hedgehog interacting protein (HHIP) is also a coreceptor, but it functions to blunt Indian hedgehog signaling by sequestering Indian hedgehog and thus preventing Indian hedgehog from binding to PTC/PTCH.


The SHH protein is modified post-translationally by the addition of cholesterol and palmitate moieties to the N- and C-termini, respectively. These lipid modifications affect SHH’s association with the cell membrane, formation of SHH multimers, and movement of SHH, altering its tissue distribution and concentration gradients. One of the best explained activities of SHH in vertebrate development is its role in patterning the ventral neural tube (see Chapters 4 and 17 ). SHH is secreted at high levels by the notochord. The concentration of SHH is highest in the floor plate of the neural tube and lowest in the roof plate, where members of the TGF-β family are highly expressed. The cell fates of four ventral interneuron classes and motor neurons are determined by relative SHH concentrations and by a combinatorial code of homeobox and basic HLH (bHLH) genes.


The requirement of SHH signaling for many developmental processes is underscored by the discovery of human mutations of members of the Shh pathway and the corresponding phenotypes of genetically modified mice, in which members are inactivated (loss of function or knockout) or overexpressed (gain of function). Mutations of SHH and PTCH have been associated with holoprosencephaly, a congenital brain defect resulting in the fusion of the two cerebral hemispheres; anophthalmia or cyclopia (see Chapter 18 ); and dorsalization of forebrain structures. In sheep, this defect can also result from exposure to the teratogen cyclopamine, which disrupts SHH signaling (see Fig. 21.5 ). Some patients with severe forms of the inborn error of cholesterol synthesis, the autosomal recessive Smith–Lemli–Opitz syndrome, have holoprosencephaly (see Chapter 20 ).


GLI3 mutations are associated with autosomal dominant polydactyly syndromes (see Chapter 16 ), such as the Greig and Pallister–Hall syndromes. Gorlin syndrome, which often is caused by germline PTCH mutations, is a constellation of congenital malformations mostly affecting the epidermis, craniofacial structures (see Chapter 9 ), and the nervous system. These patients are significantly predisposed to basal cell carcinomas, especially after radiation therapy, and a few develop malignant brain tumors (medulloblastomas) during childhood. Somatic mutations of PTCH , SUFU , and SMO have been identified in patients with sporadic medulloblastomas not associated with Gorlin syndrome.


In vertebrates, the Shh signaling pathway is closely linked to primary cilia (see Fig. 21.5 , inset) and their constituent intraflagellar transport (IFT) and basal body proteins. Primary cilia are sometimes referred to as nonmotile cilia. IFT proteins act upstream of the GLI activator (GLI-A) and repressor (GLI-R) proteins and are necessary for their production. Mutations involving genes encoding basal body proteins, such as KIAA0586 (formerly TALPID3) and oral-facial-digital syndrome 1 (OFD1) , affect SHH signaling in knockout mice. A group of human cilia-related diseases called ciliopathies result from disruption of primary cilia function and includes rare genetic diseases and more common disorders such as autosomal recessive polycystic kidney disease. To date, almost 40 ciliopathies have been described involving up to 200 genes. Although there may be some overlap (as with many congenital heart defects and left–right asymmetries), diseases of primary, nonmotile cilia are usually distinguished from disorders affecting motile cilia (found in sperm and in epithelial cells lining the airways, ventricles of the brain, and oviducts). Manifestations of diseases affecting motile cilia include hydrocephalus, lung infections, and infertility. Table 21.2 lists some common ciliopathies involving primary, nonmotile cilia and affected organ systems.



Table 21.2

Examples of Ciliopathies Due to Defects in Primary, Nonmotile Cilia

















































Disease Organ Systems Involved
Bardet–Biedl syndrome Multisystemic
Oral-facial-digital syndromes Multisystemic
Oculocerebrorenal syndrome of Lowe Multisystemic
Meckel–Gruber syndrome Brain, kidney, skeleton
Holoprosencephaly Brain, eye
Cone–rod dystrophy Eye
Leber congenital amaurosis Eye
Hearing loss Ear
Polycystic kidney disease Kidney
Nephronophthisis Kidney
Situs inversus Heart, organ laterality
Greig cephalopolysyndactyly syndrome Skeleton
Orofaciodigital syndrome Skeleton
Ellis van Creveld syndrome Skeleton


WNT/β-Catenin Pathway


The WNT -encoded glycoproteins are vertebrate orthologs of the Drosophila gene Wingless (Wg/DWnt) . Similar to the other morphogens previously discussed, the 19 WNT family members control several processes during development, including establishment of cell polarity, proliferation, apoptosis, cell fate specification, and migration. WNT signaling is very complex, and three signaling pathways have been elucidated. The classic or canonical β-catenin–dependent pathway is discussed here.


In mammals, specific WNTs bind to one of 10 Frizzled (FZD) seven transmembrane domain cell surface receptors and bind with low-density lipoprotein receptor–related protein (LRP5/LRP6) coreceptors, thereby activating downstream intracellular signaling events ( Fig. 21.6 ). β-Catenin plays an integral role in canonical WNT signaling. In the absence of WNT binding, in a protein complex with adenomatous polyposis coli (APC) and axin, cytoplasmic β-catenin is phosphorylated by glycogen synthase kinase 3 (GSK3) and targeted for degradation. In the presence of Wnts, GSK3 is phosphorylated by Dishevelled (DVL) and inactivated; it cannot phosphorylate β-catenin. β-Catenin is stabilized, accumulates in the cytoplasm, and translocates to the nucleus, where it activates target gene transcription in a complex with T-cell factor (TCF) transcription factors. In mammals, the many β-catenin/TCF target genes include those of vascular endothelial growth factor ( VEGF ), MYC, and matrix metalloproteinases (e.g., COMP, DMP1, ECM1 ).




Fig. 21.6


WNT/β-catenin canonical signaling pathway in mammals. A , In the absence of Wnt ligand binding to the Frizzled (FZD) receptor, β-catenin is phosphorylated (-P) by a multiprotein complex and targeted for degradation. Target gene expression is repressed by T-cell factor (TCF). B , When WNT binds to the FZD receptor, lipoprotein receptor–related protein (LRP) coreceptors are recruited, Dishevelled (DVL) is phosphorylated, and β-catenin then accumulates in the cytoplasm. Some β-catenin enters the nucleus to activate target gene transcription. APC , Adenomatous polyposis coli; GSK3 , glycogen synthase kinase 3.


Some noncanonical WNT signaling pathways include Frizzled receptors. However, all of these pathways are distinguished from the canonical WNT pathway by not involving β-catenin stabilization, degradation, and nuclear translocation. A well-known noncanonical Wnt signaling pathway is the WNT-cGMP/Ca 2+ pathway. It acts through phospholipase C (PLC) to increase intracellular calcium concentrations, thereby activating protein kinase C (PKC) and calmodulin-dependent kinase II (CamKII) and resulting in myriad downstream effects.


In mammals, dysregulated WNT signaling is a prominent feature in many developmental disorders and cancer. A Frizzled gene ( FZD9 ) occurs in the Williams–Beuren syndrome deletion region. LRP5 mutations are found in the osteoporosis-pseudoglioma syndrome. Dvl2 -knockout mice have malformations of the cardiac outflow tract, abnormal somite segmentation, and neural tube defects. Similar to the Shh signaling pathway, canonical Wnt pathway mutations (in β -catenin [ CTNNB1 ], APC , and AXIN1 genes) have been described in children with medulloblastoma. Moreover, somatic APC mutations are common (>50%) in adults with sporadic colorectal carcinomas, and germline APC mutations are a feature of familial adenomatous polyposis and Turcot syndrome (multiple colorectal adenomas and increased frequency of primary brain tumors).




Protein Kinases


Receptor Tyrosine Kinases


Common Features


Growth factors include insulin , epidermal growth factor, nerve growth factor and other neurotrophins, and members of the platelet-derived growth factor family. Growth factors bind to cell surface transmembrane receptors found on target cells. These receptors are members of the RTK superfamily and have three domains: an extracellular ligand binding domain, a transmembrane domain, and an intracellular kinase domain ( Fig. 21.7 ).




Fig. 21.7


Receptor tyrosine kinase (RTK) signaling. A , In the absence of ligand, the receptors are monomers and are inactive. B , On binding of ligand, the receptors dimerize and transphosphorylation occurs, which activates downstream signaling cascades. P , Phosphorylated.


The receptors are found as monomers in the quiescent or unbound state, but upon ligand binding, the receptor units dimerize. The process of dimerization brings the two intracellular kinase domains close enough that one kinase domain can phosphorylate and activate the other receptor (transphosphorylation). Transphosphorylation is required to fully activate the receptors, which initiate a series of intracellular signaling cascades. The mechanism of transphosphorylation requires both receptor subunits within a dimer to have functional kinase domains for signal transduction to occur. If there is an inactivating mutation of one receptor subunit’s kinase domain, the functional consequence is to abolish signaling through a heterodimer resulting from the combination of wild-type and mutant receptor subunits (dominant negative mode of action). A mutation in the kinase domain of the VEGF receptor 3 (VEGFR3, now called FMS-related tyrosine kinase 4 [FLT4]) results in the autosomal dominant lymphatic disorder called Milroy disease.


Regulation of Angiogenesis by Receptor Tyrosine Kinases


Growth factors promote cellular proliferation, migration, and survival; they are antiapoptotic. Dysregulation of RTKs or their downstream signaling components is frequently found in human cancers. During embryogenesis, signaling through RTK is crucial for normal development and affects many processes, such as the growth of new blood vessels (see Chapter 4 ), cellular migration, and neuronal axonal guidance.


Endothelial cells are derived from a progenitor cell (hemangioblast) that can give rise to the hematopoietic cell lineage and endothelial cells. The early endothelial cells proliferate and eventually coalesce to form the first primitive blood vessels. This process is called vasculogenesis . After the first blood vessels are formed, they undergo intensive remodeling and maturation into the mature blood vessels in a process called angiogenesis . The maturation process involves the recruitment of vascular smooth muscle cells to the vessels that stabilize them. Vasculogenesis and angiogenesis depend on the function of two distinct RTK classes, members of the VEGF and TIE (tyrosine kinase with immunoglobulin-like and EGF-like domains) receptor families. VEGFA is essential for endothelial and blood cell development. VegfA -knockout mice fail to develop blood or endothelial cells and die at early embryonic stages. Heterozygous VegfA-knockout mice have severe defects in their vasculature, demonstrating that VegfA gene dose (haploinsufficiency) is important. A related molecule, VEGFC, is crucial for the development of lymphatic endothelial cells. VEGFA signals through two receptors, VEGFR1 and VEGFR2, expressed by endothelial cells. VEGFA signals predominantly through the VEGFR2 receptor for vasculogenesis to properly occur in the embryo.


The process of angiogenic refinement depends on the function of the Angiopoietin/TIE2 signaling pathway. TIE2 (also called TEK) is an RTK that is specifically expressed by endothelial cells, and Angiopoietin 1 and Angiopoietin 2 are its ligands that are expressed by the surrounding vascular smooth muscle cells. This represents a paracrine signaling system in which receptor and ligand are expressed in adjacent cells. The VEGF/VEGFR2 and Angiopoietin/TIE2 signaling pathways are co-opted by tumors to stimulate the growth of new blood vessels, which stimulates their growth and metastasis. This demonstrates how normal signaling pathways in the developing human can be reused for disease processes such as cancer.


Hippo Signaling Pathway


Studies in Drosophila identified a set of kinases in the Hippo signaling pathway that, when mutated, resulted in increased organ size during development. The human orthologs of Hippo are called mammalian STE20-like protein kinase 1 (MST1) and MST2. Activated MST1 and MST2 phosphorylate the scaffold protein Salvador homolog 1 (SAV1) and the downstream kinases large tumor suppressor homolog 1 (LATS1) and LATS2 ( Fig. 21.8 ). Similar to MST1 and MST2, LATS1 and LATS2 are bound to scaffold proteins MOB kinase activator 1A (MOB1A) and MOB1B, which are also phosphorylated by MST1 and MST2. The MST/MOB1 complex then phosphorylates the transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ domain (TAZ). Phosphorylated YAP and TAZ proteins are retained in the cytoplasm, ubiquitinated, and degraded by the proteasome.


Mar 31, 2020 | Posted by in GENERAL | Comments Off on Common Signaling Pathways Used During Development

Full access? Get Clinical Tree

Get Clinical Tree app for offline access