Uncommon Descent Serving The Intelligent Design Community

The Ubiquitin System: Functional Complexity and Semiosis joined together.

Share
Facebook
Twitter
LinkedIn
Flipboard
Print
Email

This is a very complex subject, so as usual I will try to stick to the essentials to make things as clear as possible, while details can be dealt with in the discussion.

It is difficult to define exactly the role of the Ubiquitin System. It is usually considered mainly a pathway which regulates protein degradation, but in reality its functions are much wider than that.

In essence, the US is a complex biological system which targets many different types of proteins for different final fates.

The most common “fate” is degradation of the protein. In that sense, the Ubiquitin System works together with another extremely complex cellular system, the proteasome. In brief, the Ubiquitin System “marks” proteins for degradation, and the proteasome degrades them.

It seems simple. It is not.

Ubiquitination is essentially one of many Post-Translational modifications (PTMs): modifications of proteins after their synthesis by the ribosome (translation). But, while most PTMs use simpler biochemical groups that are usually added to the target protein (for example, acetylation), in ubiquitination a whole protein (ubiquitin) is used as a modifier of the target protein.

 

The tool: Ubiquitin

Ubiquitin is a small protein (76 AAs). Its name derives from the simple fact that it  is found in most tissues of eukaryotic organisms.

Here is its aminoacid sequence:

MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPD

QQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG

Essentially, it has two important properties:

  1. As said, it is ubiquitous in eukaryotes
  2. It is also extremely conserved in eukaryotes

In mammals, ubiquitin is not present as a single gene. It is encoded by 4 different genes: UBB, a poliubiquitin (3 Ub sequences); UBC, a poliubiquitin (9 Ub sequences); UBA52, a mixed gene (1   Ub sequence + the ribosomal protein L40); and RPS27A, again a mixed gene (1 Ub sequence + the ribosomal protein S27A). However, the basic ubiquitin sequence is always the same in all those genes.

Its conservation is one of the highest in eukaryotes. The human sequence shows, in single celled eukaryotes:

Naegleria: 96% conservation;  Alveolata: 100% conservation;  Cellular slime molds: 99% conservation; Green algae: 100% conservation; Fungi: best hit 100% conservation (96% in yeast).

Ubiquitin and Ubiquitin like proteins (see later) are characterized by a special fold, called  β-grasp fold.

 

The semiosis: the ubiquitin code

The title of this OP makes explicit reference to semiosis. Let’s try to see why.

The simplest way to say it is: ubiquitin is a tag. The addition of ubiquitin to a substrate protein marks that protein for specific fates, the most common being degradation by the proteasome.

But not only that. See, for example, the following review:

Nonproteolytic Functions of Ubiquitin in Cell Signaling

Abstract:

The small protein ubiquitin is a central regulator of a cell’s life and death. Ubiquitin is best known for targeting protein destruction by the 26S proteasome. In the past few years, however, nonproteolytic functions of ubiquitin have been uncovered at a rapid pace. These functions include membrane trafficking, protein kinase activation, DNA repair, and chromatin dynamics. A common mechanism underlying these functions is that ubiquitin, or polyubiquitin chains, serves as a signal to recruit proteins harboring ubiquitin-binding domains, thereby bringing together ubiquitinated proteins and ubiquitin receptors to execute specific biological functions. Recent advances in understanding ubiquitination in protein kinase activation and DNA repair are discussed to illustrate the nonproteolytic functions of ubiquitin in cell signaling.

Another important aspect is that ubiquitin is not one tag, but rather a collection of different tags. IOWs, a tag based code.

See, for example, here:

The Ubiquitin Code in the Ubiquitin-Proteasome System and Autophagy

(Paywall).

Abstract:

The conjugation of the 76 amino acid protein ubiquitin to other proteins can alter the metabolic stability or non-proteolytic functions of the substrate. Once attached to a substrate (monoubiquitination), ubiquitin can itself be ubiquitinated on any of its seven lysine (Lys) residues or its N-terminal methionine (Met1). A single ubiquitin polymer may contain mixed linkages and/or two or more branches. In addition, ubiquitin can be conjugated with ubiquitin-like modifiers such as SUMO or small molecules such as phosphate. The diverse ways to assemble ubiquitin chains provide countless means to modulate biological processes. We overview here the complexity of the ubiquitin code, with an emphasis on the emerging role of linkage-specific degradation signals (degrons) in the ubiquitin-proteasome system (UPS) and the autophagy-lysosome system (hereafter autophagy).

A good review of the basics of the ubiquitin code can be found here:

The Ubiquitin Code 

(Paywall)

It is particularly relevant, from an ID point of view, to quote the starting paragraph of that paper:

When in 1532 Spanish conquistadores set foot on the Inca Empire, they found a highly organized society that did not utilize a system of writing. Instead, the Incas recorded tax payments or mythology with quipus, devices in which pieces of thread were connected through specific knots. Although the quipus have not been fully deciphered, it is thought that the knots between threads encode most of the quipus’ content. Intriguingly, cells use a regulatory mechanism—ubiquitylation—that is reminiscent of quipus: During this reaction, proteins are modified with polymeric chains in which the linkage between ubiquitin molecules encodes information about the substrate’s fate in the cell.

Now, ubiquitin is usually linked to the target protein in chains. The first ubiquitin molecule is covalently bound through its C-terminal carboxylate group to a particular lysine, cysteine, serine, threonine or N-terminus of the target protein.

Then, additional ubiquitins are added to form a chain, and the C-terminus of the new ubiquitin is linked to one of seven lysine residues or the first methionine residue on the previously added ubiquitin.

IOWs, each ubiquitin molecule has seven lysine residues:

K6, K11, K27, K29, K33, K48, K63

And one N terminal methionine residue:

M1

And a new ubiquitin molecule can be added at each of those 8 sites in the previous ubiquitin molecule. IOWs, those 8 sites in the molecule are configurable switches that can be used to build ubiquitin chains.

Her are the 8 sites, in red, in the ubiquitin molecule:

MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPD

QQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG

Fig 1 shows two ubiquitin molecules joined at K48.

Fig 1 A cartoon representation of a lysine 48-linked diubiquitin molecule. The two ubiquitin chains are shown as green cartoons with each chain labelled. The components of the linkage are indicated and shown as orange sticks. By Rogerdodd (Own work) [CC BY-SA 3.0 (https://creativecommons.org/licenses/by-sa/3.0)], via Wikimedia Commons

The simplest type of chain is homogeneous (IOWs, ubiquitins are linked always at the same site). But many types of mixed and branched chains can also be found.

Let’s start with the most common situation: a poli-ubiquitination of (at least) 4 ubiqutins, linearly linked at K48. This is the common signal for proteasome degradation.

By the way, the 26S proteasome is another molecular machine of incredible complexity, made of more than 30 different proteins. However, its structure and function are not the object of this OP, and therefore I will not deal with them here.

The ubiquitin code is not completely understood, at present, but a few aspects have been well elucidated. Table 1 sums up the most important and well known modes:

Code

Meaning

Polyubiquitination (4 or more) with links at K48 or at K11 Proteasomal degradation
Monoubiqutination (single or multiple) Protein interactions, membrane trafficking, endocytosis
Polyubiquitination with links at K63 Endocytic trafficking, inflammation, translation, DNA repair.
Polyubiquitination with links at K63 (other links) Autophagic degradation of protein substrates
Polyubiquitination with links at K27, K29, K33 Non proteolytic processes
Rarer chain types (K6, K11) Under investigation

 

However, this is only a very partial approach. A recent bioinformatics paper:

An Interaction Landscape of Ubiquitin Signaling

(Paywall)

Has attempted for the first time a systematic approach to deciphering the whole code, using synthetic diubiquitins (all 8 possible variants) to identify the different interactors with those signals, and they identified, with two different methodologies,  111 and 53 selective interactors for linear polyUb chains, respectively. 46 of those interactors were identified by both methodologies.

The translation

But what “translates” the complex ubiquitin code, allowing ubiquinated proteins to met the right specific destiny? Again, we can refer to the diubiquitin paper quoted above.

How do cells decode this ubiquitin code into proper cellular responses? Recent studies have indicated that members of a protein family, ubiquitin-binding proteins (UBPs), mediate the recognition of ubiquitinated substrates. UBPs contain at least one of 20 ubiquitin-binding domains (UBDs) functioning as a signal adaptor to transmit the signal from ubiquitinated substrates to downstream effectors

But what are those “interactors” identified by the paper (at least 46 of them)? They are, indeed, complex proteins which recognize specific configurations of the “tag” (the ubiquitin chain), and link the tagged (ubiquinated) protein to other effector proteins which implement its final fate, or anyway contribute in deffrent forms to that final outcome.

 

The basic control of the procedure: the complexity of the ubiquitination process.

So, we have seen that ubiquitin chains work as tags, and that their coded signals are translated by specific interactors, so that the target protein may be linked to its final destiny, or contribute to the desired outcome. But we must still address one question: how is the ubiquitination of the different target proteins implemented? IOWs, what is the procedure that “writes” the specific codes associated to specific target proteins?

This is indeed the first step in the whole process. But it is also the most complex, and that’s why I have left it for the final part of the discussion.

Indeed, the ubiquitination process needs to realize the following aims:

  1. Identify the specific protein to be ubiquitinated
  2. Recognize the specific context in which that protein needs to be ubiquitinated
  3. Mark the target protein with the correct tag for the required fate or outcome

We have already seen that the ubiquitin system is involved in practically all different cellular paths and activities, and therefore we can expect that the implementation of the above functions must be a very complex thing.

And it is.

Now, we can certainly imagine that there are many different layers of regulation that may contribute to the general control of the procedure, specifically epigenetic levels, which are at present poorly understood. But there is one level that we can more easily explore and understand, and it is , as usual, the functional complexity of the proteins involved.

And, even at a first gross analysis, it is really easy to see that the functional complexity implied by this process is mind blowing.

Why? It is more than enough to consider the huge number of different proteins involved. Let’s see.

The ubiquitination process is well studied. It can be divided into three phases, each of which is implemented by a different kind of protein. The three steps, and the three kinds of proteins that implement them, take the name of E1, E2 and E3.

 

Fig. 2 Schematic diagram of the ubiquitylation system. Created by Roger B. Dodd: Rogerdodd at the English language Wikipedia [GFDL (http://www.gnu.org/copyleft/fdl.html) or CC-BY-SA-3.0 (http://creativecommons.org/licenses/by-sa/3.0/)], via Wikimedia Commons

 The E1 step of ubiquitination.

This is the first thing that happens, and it is also the simplest.

E1 is the process of activation of ubiquitin, and the E1 proteins is called E1 ubiquitin-activating enzyme. To put it simply, this enzyme “activates” the ubiquitin molecule in an ATP dependent process, preparing it for the following phases and attaching it to its active site cysteine residue. It is not really so simple, but for our purposes that can be enough.

This is a rather straightforward enzymatic reaction. In humans there are essentially two forms of E1 enzymes, UBA1 and UBA6, each of them about 1000 AAs long, and partially related at sequence level (42%).

 

The E2 step of ubiquitination.

The second step is ubiquitin conjugation. The activated ubiquitin is transferred from the E1 enzyme to the ubiquitin-conjugating enzyme, or E2 enzyme, where it is attached to a cysteine residue.

This apparently simple “transfer” is indeed a complex intermediate phase. Humans have about 40 different E2 molecules. The following paper:

E2 enzymes: more than just middle men

details some of the functional complexity existing at this level.

Abstract:

Ubiquitin-conjugating enzymes (E2s) are the central players in the trio of enzymes responsible for the attachment of ubiquitin (Ub) to cellular proteins. Humans have ∼40 E2s that are involved in the transfer of Ub or Ub-like (Ubl) proteins (e.g., SUMO and NEDD8). Although the majority of E2s are only twice the size of Ub, this remarkable family of enzymes performs a variety of functional roles. In this review, we summarize common functional and structural features that define unifying themes among E2s and highlight emerging concepts in the mechanism and regulation of E2s.

However, I will not go into details about these aspects, because we have better things to do: we still have to discuss the E3 phase!

 

The E3 step of ubiquitination.

This is the last phase of ubiquitination, where the ubiquitin tag is finally transferred to the target protein, as initial mono-ubiquitination, or to build an ubiquitin chain by following ubiqutination events. The proteins which implement this final passage are call E3 ubiquitin ligases. Here is the definition from Wikipedia:

A ubiquitin ligase (also called an E3 ubiquitin ligase) is a protein that recruits an E2 ubiquitin-conjugating enzyme that has been loaded with ubiquitin, recognizes a protein substrate, and assists or directly catalyzes the transfer of ubiquitin from the E2 to the protein substrate.

It is rather obvious that the role of the E3 protein is very important and delicate. Indeed it:

  1. Recognizes and links the E2-ubiquitin complex
  2. Recognizes and links some specific target protein
  3. Builds the appropriate tag for that protein (Monoubiquitination, mulptiple monoubiquitination, or poliubiquitination with the appropriate type of ubiquitin chain).
  4. And it does all those things at the right moment, in the right context, and for the right protein.

IOWs, the E3 protein writes the coded tag. It is, by all means, the central actor in our complex story.

So, here comes the really important point: how many different E3 ubiquitin ligases do we find in eukaryotic organisms? And the simple answer is: quite a lot!

Humans are supposed to have more than 600 different E3 ubiquitin ligases!

So, the human machinery for ubiquitination is about:

2 E1 proteins  –  40 E2 proteins – >600 E3 proteins

A real cascade of complexity!

OK, but even if we look at single celled eukaryotes we can already find an amazing level of complexity. In yeast, for example, we have:

1 or 2 E1 proteins  –  11 E2 proteins – 60-100 E3 proteins

See here:

The Ubiquitin–Proteasome System of Saccharomyces cerevisiae

Now, a very important point. Those 600+ E3 proteins that we find in humans are really different proteins. Of course, they have something in common: a specific domain.

From that point of view, they can be roughly classified in three groups according to the specific E3 domain:

  1. RING group: the RING finger domain ((Really Interesting New Gene) is a short domain of zinc finger type, usually 40 to 60 amino acids. This is the biggest group of E3s (about 600)
  2. HECT domain (homologous to the E6AP carboxyl terminus): this is a bigger domain (about 350 AAs). Located at the C terminus of the protein. It has a specific ligase activity, different from the RING   In humans we have approximately 30 proteins of this type.
  3. RBR domain (ring between ring fingers): this is a common domain (about 150 AAs) where two RING fingers are separated by a region called IBR, a cysteine-rich zinc finger. Only a subset of these proteins are E3 ligases, in humans we have about 12 of them.

See also here.

OK, so these proteins have one of these three domains in common, usually the RING domain. The function of the domain is specifically to interact with the E2-ubiquitin complex to implement the ligase activity. But the domain is only a part of the molecule, indeed a small part of it. E3 ligases are usually big proteins (hundreds, and up to thousands of AAs). Each of these proteins has a very specific non domain sequence, which is probably responsible for the most important part of the function: the recognition of the specific proteins that each E3 ligase processes.

This is a huge complexity, in terms of functional information at sequence level.

Our map of the ubiquinating system in humans could now be summarized as follows:

2 E1 proteins  –  40 E2 proteins – 600+ E3 proteins + thousands of specific substrates

IOWs, each of hundreds of different complex proteins recognizes its specific substrates, and marks them with a shared symbolic code based on uniquitin and its many possible chains. And the result of that process is that proteins are destined to degradation by the proteasome or other mechanisms, and that protein interactions and protein signaling are regulated and made possible, and that practically all cellular functions are allowed to flow correctly and smoothly.

Finally, here are two further compoments of the ubuquitination system, which I will barely mention, to avoid making this OP too long.

Ubiquitin like proteins (Ubl):

A number of ubiquitin like proteins add to the complexity of the system. Here is the abstract from a review:

The eukaryotic ubiquitin family encompasses nearly 20 proteins that are involved in the posttranslational modification of various macromolecules. The ubiquitin-like proteins (UBLs) that are part of this family adopt the β-grasp fold that is characteristic of its founding member ubiquitin (Ub). Although structurally related, UBLs regulate a strikingly diverse set of cellular processes, including nuclear transport, proteolysis, translation, autophagy, and antiviral pathways. New UBL substrates continue to be identified and further expand the functional diversity of UBL pathways in cellular homeostasis and physiology. Here, we review recent findings on such novel substrates, mechanisms, and functions of UBLs.

These proteins include SUMO, Nedd8, ISB15, and many others.

Deubiquitinating enzymes (DUBs):

The process of ubiquitination, complex as it already is, is additionally regulated by these enzymes which can cleave ubiquitin from proteins and other molecules. Doing so, they can reverse the effects of ubiquitination, creating a delicately balanced regulatory network. In humans there are nearly 100 DUB genes, which can be classified into two main classes: cysteine proteases and metalloproteases.

 

By the way, here is a beautiful animation of the basic working of the ubiquitin-proteasome system in degrading damaged proteins:

 

 

A summary:

So, let’s try a final graphic summary of the whole ubiquitin system in humans:

Fig 3 A graphic summary of the Ubiquitin System

 

Evolution of the Ubiquitin system?

The Ubiqutin system is essentially an eukaryotic tool. Of course, distant precursors for some of the main components have been “found” in prokaryotes. Here is the abstract from a paper that sums up what is known about the prokaryotic “origins” of the system:

Structure and evolution of ubiquitin and ubiquitin-related domains.

(Paywall)

Abstract:

Since its discovery over three decades ago, it has become abundantly clear that the ubiquitin (Ub) system is a quintessential feature of all aspects of eukaryotic biology. At the heart of the system lies the conjugation and deconjugation of Ub and Ub-like (Ubls) proteins to proteins or lipids drastically altering the biochemistry of the targeted molecules. In particular, it represents the primary mechanism by which protein stability is regulated in eukaryotes. Ub/Ubls are typified by the β-grasp fold (β-GF) that has additionally been recruited for a strikingly diverse range of biochemical functions. These include catalytic roles (e.g., NUDIX phosphohydrolases), scaffolding of iron-sulfur clusters, binding of RNA and other biomolecules such as co-factors, sulfur transfer in biosynthesis of diverse metabolites, and as mediators of key protein-protein interactions in practically every conceivable cellular context. In this chapter, we present a synthetic overview of the structure, evolution, and natural classification of Ub, Ubls, and other members of the β-GF. The β-GF appears to have differentiated into at least seven clades by the time of the last universal common ancestor of all extant organisms, encompassing much of the structural diversity observed in extant versions. The β-GF appears to have first emerged in the context of translation-related RNA-interactions and subsequently exploded to occupy various functional niches. Most biochemical diversification of the fold occurred in prokaryotes, with the eukaryotic phase of its evolution mainly marked by the expansion of the Ubl clade of the β-GF. Consequently, at least 70 distinct Ubl families are distributed across eukaryotes, of which nearly 20 families were already present in the eukaryotic common ancestor. These included multiple protein and one lipid conjugated forms and versions that functions as adapter domains in multimodule polypeptides. The early diversification of the Ubl families in eukaryotes played a major role in the emergence of characteristic eukaryotic cellular substructures and systems pertaining to nucleo-cytoplasmic compartmentalization, vesicular trafficking, lysosomal targeting, protein processing in the endoplasmic reticulum, and chromatin dynamics. Recent results from comparative genomics indicate that precursors of the eukaryotic Ub-system were already present in prokaryotes. The most basic versions are those combining an Ubl and an E1-like enzyme involved in metabolic pathways related to metallopterin, thiamine, cysteine, siderophore and perhaps modified base biosynthesis. Some of these versions also appear to have given rise to simple protein-tagging systems such as Sampylation in archaea and Urmylation in eukaryotes. However, other prokaryotic systems with Ubls of the YukD and other families, including one very close to Ub itself, developed additional elements that more closely resemble the eukaryotic state in possessing an E2, a RING-type E3, or both of these components. Additionally, prokaryotes have evolved conjugation systems that are independent of Ub ligases, such as the Pup system.

 

As usual, we are dealing here with distant similarities, but there is no doubt that the ubiquitin system as we know it appears in eukaryotes.

But what about its evolutionary history in eukaryotes?

We have already mentioned the extremely high conservation of ubiquitin itself.

UBA1, the main E1 enzyme, is rather well conserved from fungi to humans: 60% identity, 1282 bits, 1.21 bits per aminoacid (baa).

E2s are small enzymes, extremely conserved from fungi to humans: 86% identity, for example, for UB2D2, a 147 AAs molecule.

E3s, of course, are the most interesting issue. This big family of proteins behaves in different ways, consistently with its highly specific functions.

It is difficult to build a complete list of E3 proteins. I have downloaded from Uniprot a list of reviewed human proteins including “E3 ubiquitun ligase” in their name: a total of 223 proteins.

The mean evolutionary behavior of this group in metazoa is rather different from protein to protein. However, as a group these proteins exhibit an information jump in vertebrates which is significantly higher than the jump in all other proteins:

 

Fig. 4 Boxplots of the distribution of human conserved information jump from pre-vertebrates to vertebrates in 223 E3 ligase proteins and in all other human proteins. The difference is highly significant.

 

As we already know, this is evidence that this class of proteins is highly engineered in the transition to vertebrates. That is consistent with the need to finely regulate many cellular processes, most of which are certainly highly specific for different groups of organisms.

The highest vertebrate jump, in terms of bits per aminoacid, is shown in my group by the E3 ligase TRIM62. also known as DEAR1 (Q9BVG3), a 475 AAs long protein almost absent in pre-vertebrates (best hit 129 bits, 0.27 baa in Branchiostoma belcheri) and which flaunts an amazing jump of 1.433684 baa in cartilaginous fish (810 bits, 1.705263 baa).

But what is this protein? It is a master regulator tumor suppressor gene, implied in immunity, inflammation, tumor genesis.

See here:

TRIM Protein-Mediated Regulation of Inflammatory and Innate Immune Signaling and Its Association with Antiretroviral Activity

and here:

DEAR1 is a Chromosome 1p35 Tumor Suppressor and Master Regulator of TGFβ-Driven Epithelial-Mesenchymal Transition

This is just to show what a single E3 ligase can be involved in!

An opposite example, from the point of view of evolutionary history, is SIAH1, an E3 ligase implied in proteosomal degradation of proteins. It is a 282 AAs long protein, which already exhibits 1.787234 baa (504 bits) of homology in deuterostomes, indeed already 1.719858 baa in cnidaria. However, in fungi the best hit is only 50.8 bits (0.18 baa). So, this is a protein whose engineering takes place at the start of metazoa, and which exhibits only a minor further jump in vertebrates (0.29 baa), which brings the protein practically to its human form already in cartilaginous fish (280 identities out of 282, 99%). Practically a record.

So, we can see that E3 ligases are a good example of a class of proteins which perform different specific functions, and therefore exhibit different evolutionary histories: some, like TRIM62, are vertebrate quasi-novelties, others, like SIAH1, are metazoan quasi-novelties. And, of course, there are other behaviours, like for example BRCA1, Breast cancer type 1 susceptibility protein, a protein 1863 AAs long which only in mammals acquires part of its final sequence configuration in humans.

The following figure shows the evolutionary history of the three proteins mentioned above.

 

Fig. 5 Evolutionary history in metazoa of three E3 ligases (human conserved functional information)

 

An interesting example: NF-kB signaling

I will discuss briefly an example of how the Ubiquitin system interacts with some specific and complex final effector system. One of the best models for that is the NF-kB signaling.

NK-kB is a transcription factor family that is the final effector of a complex signaling pathway. I will rely mainly on the following recent free paper:

The Ubiquitination of NF-κB Subunits in the Control of Transcription

Here is the abstract:

Nuclear factor (NF)-κB has evolved as a latent, inducible family of transcription factors fundamental in the control of the inflammatory response. The transcription of hundreds of genes involved in inflammation and immune homeostasis require NF-κB, necessitating the need for its strict control. The inducible ubiquitination and proteasomal degradation of the cytoplasmic inhibitor of κB (IκB) proteins promotes the nuclear translocation and transcriptional activity of NF-κB. More recently, an additional role for ubiquitination in the regulation of NF-κB activity has been identified. In this case, the ubiquitination and degradation of the NF-κB subunits themselves plays a critical role in the termination of NF-κB activity and the associated transcriptional response. While there is still much to discover, a number of NF-κB ubiquitin ligases and deubiquitinases have now been identified which coordinate to regulate the NF-κB transcriptional response. This review will focus the regulation of NF-κB subunits by ubiquitination, the key regulatory components and their impact on NF-κB directed transcription.

 

The following figure sums up the main features of the canonical activation pathway:

 

Fig. 6 A simple summary of the main steps in the canonical activayion pathway of NF-kB

 

Here the NF-κB TF is essentially the heterodimer RelA – p50. Before activation, the NF-κB (RelA – p50) dimer is kept in an inactive state and remains in the cytoplasm because it is linked to the IkB alpha protein, an inhibitor of its function.

Activation is mediated by a signal-receptor interaction, which starts the whole pathway. A lot of different signals can do that, adding to the complexity, but we will not discuss this part here.

As a consequence of receptor activation, another protein complex, IκB kinase (IKK), accomplishes the Phosphorylation of IκBα at serines 32 and 36. This is the signal for the ubiquitination of the IkB alpha inhibitor.

This ubiqutination targets IkB alpha for proteosomal degradation. But how is it achieved?

Well, things are not so simple. A whole protein complex is necessary, a complex which implements many different ubiquitinations in different contexts, including this one.

The complex is made by 3 basic proteins:

  • Cul1 (a scaffold protein, 776 AAs)
  • SKP1 (an adaptor protein, 163 AAs)
  • Rbx1 (a RING finger protein with E3 ligase activity, 108 AAs)

Plus:

  • An F-box protein (FBP) which changes in the different context, and confers specificity.

In our context, the F box protein is called beta TRC (605 AAs).

 

Fig. 7 A simple diagram of the SKP1 – beta TRC complex

 

Once the IkB alpha inhibitor is ubiquinated and degraded in the proteasome, the NF-κB dimer is free to translocate to the nucleus, and implement its function as a transcription factor (which is another complex issue, that we will not discuss).

OK, this is only the canonical activation of the pathway.

In the non canonical pathway (not shown in the figure) a different set of signals, receptors and activators acts on a different NF-κB dimer (RelB – p100). This dimer is not linked to any inhibitor, but is itself inactive in the cytoplasm. As a result of the signal, p100 is phosphorylated at serines 866 and 870. Again, this is the signal for ubiquitination.

This ubiquitination is performed by the same complex described above, but the result is different. P100 is only partially degraded in the proteasome, and is transformed into a smaller protein, p52, which remains linked to RelB. The RelB – p52 dimer is now an active NF-κB Transcription Factor, and it can relocate to the nucleus and act there.

But that’s not all.

  • You may remember that RelA (also called p 65) is one of the two components of NF-kB TF in the canonical pathway (the other being p 50). Well, RelA is heavily controlled by ubiquitination after it binds DNA in the nucleus to implement its TF activity. Ubiquitination (a very complex form of it) helps detachment of the TF from DNA, and its controlled degradation, avoiding sustained expression of NF-κB-dependent genes. For more details, see section 4 in the above quoted paper: “Ubiquitination of NF-κB”.
  • The activation of IKK in both the canonical and non canonical pathway after signal – receptor interaction is not so simple as depicted in Fig. 6. For more details, look at Fig. 1 in this paper: Ubiquitin Signaling in the NF-κB Pathway. You can see that, in the canonical pathway, the activation of IKK is mediated by many proteins, including TRAF2, TRAF6, TAK1, NEMO.
  • TRAF2 is a key regulator on many signaling pathways, including NF-kB. It is an E3 ubiquitin ligase. From Uniprot:  “Has E3 ubiquitin-protein ligase activity and promotes ‘Lys-63’-linked ubiquitination of target proteins, such as BIRC3, RIPK1 and TICAM1. Is an essential constituent of several E3 ubiquitin-protein ligase complexes, where it promotes the ubiquitination of target proteins by bringing them into contact with other E3 ubiquitin ligases.”
  • The same is true of TRAF6.
  • NEMO (NF-kappa-B essential modulator ) is also a key regulator. It is not an ubiquinating enzyme, but it is rather heavily regulated by ubiquitination. From Uniprot: “Regulatory subunit of the IKK core complex which phosphorylates inhibitors of NF-kappa-B thus leading to the dissociation of the inhibitor/NF-kappa-B complex and ultimately the degradation of the inhibitor. Its binding to scaffolding polyubiquitin seems to play a role in IKK activation by multiple signaling receptor pathways. However, the specific type of polyubiquitin recognized upon cell stimulation (either ‘Lys-63’-linked or linear polyubiquitin) and its functional importance is reported conflictingly.”
  • In the non canonical pathway, the activation of IKK alpha after signal – receptor interaction is mediated by other proteins, in particular one protein called NIK (see again Fig. 1 quoted above). Well, NIK is regulated by two different types of E3 ligases, with two different types of polyubiquitination:
    • cIAP E3 ligase inactivates it by constant degradation using a K48 chain
    • ZFP91 E3 ligase stabilizes it using a K63 chain

See here:

Non-canonical NF-κB signaling pathway.

In particular, Fig. 3

These are only some of the ways the ubiquitin system interacts with the very complex NF-kB signaling system. I hope that’s enough to show how two completely different and complex biological systems manage to cooperate by intricate multiple connections, and how the ubiquitin system can intervene at all levels of another process. What is true for the NF-kB signaling pathway is equally true for a lot of other biological systems, indeed for almost all basic cellular processes.

But this OP is already too long, and I have to stop here.

As usual, I want to close with a brief summary of the main points:

  1. The Ubiquitin system is a very important regulation network that shows two different signatures of design: amazing complexity and an articulated semiotic structure.
  2. The complexity is obvious at all levels of the network, but is especially amazing at the level of the hundreds of E3 ligases, that can recognize thousands of different substrates in different contexts.
  3. The semiosis is obvious in the Ubiquitin Code, a symbolic code of different ubiquitin configurations which serve as specific “tags” that point to different outcomes.
  4. The code is universally implemented and shared in eukaryotes, and allows control on almost all most important cellular processes.
  5. The code is written by the hundreds of E3 ligases. It is read by the many interactors with ubiquitin-binding domains (UBDs).
  6. The final outcome is of different types, including degradation, endocytosis, protein signaling, and so on.
  7. The interaction of the Ubiquitin System with other complex cellular pathways, like signaling pathways, is extremely complex and various, and happens at many different levels and by many different interacting proteins for each single pathway.

PS:

Thanks to DATCG for pointing to this video in three parts by Dr. Raymond Deshaies, was Professor of Biology at the California Institute of Technology and an Investigator of the Howard Hughes Medical Institute. On iBiology Youtube page:

A primer on the ubiquitin-proteasome system

 

Cullin-RING ubiquitin ligases: structure, structure, mechanism, and regulation

 

Targeting the ubiquitin-proteasome system in cancer

Comments
Probability arguments are used for the mere fact tat there isn't any evidence that natural selection and drift could produce the biological structure in question. There isn't even a methodology to test the claim. Right, evolution is not goal oriented which makes the problems worse. Your position doesn't even deserve a seat at the probability tableET
April 11, 2018
April
04
Apr
11
11
2018
09:01 AM
9
09
01
AM
PDT
Bill Cole and Gpuccio, there are many avenues that can be used to support ID but using the combinatorial explosion is just using a probability argument to knock over a strawman view of what evolution is. The probability arguments being used would be perfectly valid if evolution was goal oriented. For example, if the goal of evolution was to produce ATP synthase, or lactase, then using the probability calculations commonly thrown around to criticize evolution would be valid. But evolution is not goal oriented like this. Just as the unique DNA sequence that is Gpuccio was not the goal (nothing personal :) ). Given his name, I am assuming that he is of Italian descent. Starting at the time of the Romans, and Gpuccio's hypothesized great-great^?? grandparents, what is the probability that Gpuccio with his unique DNA sequence would be sitting at his computer typing a response to my comment? I think that we would all agree that it would be astronomically improbable. However, what is the probability that these same two people would have extant descendants? These probability arguments assume that existing proteins and existing metabolic pathways are the only ones that were ever possible. This is not the case as is demonstrated by the number of variations on the theme observed in extant organisms.Allan Keith
April 11, 2018
April
04
Apr
11
11
2018
08:41 AM
8
08
41
AM
PDT
The trick of the argument is equivocating between a specification obtained from the results and a specification obtained independently from the results. In GPuccio's example (#859), the chance to get a result and to get a specification from that result = 1 (100%). However, the chance to get a result that matches a specification obtained independently from the results = 10^-150Origenes
April 11, 2018
April
04
Apr
11
11
2018
06:59 AM
6
06
59
AM
PDT
gpuccio:
You are really proposing again what I call “the infamous deck of cards fallacy”. One of the worst and most arrogant wrong arguments that I have ever heard.
I called it a brain-dead argument.ET
April 11, 2018
April
04
Apr
11
11
2018
06:44 AM
6
06
44
AM
PDT
Allan Keith at #850: b) Second point:
You have a very unique and specific DNA sequence. What is the probability of this arising? Your father produced millions of sperm cells, your mother thousands of ova. To create you, the exact two would have had to get together. Add to this the probability of your parents getting together at the right time (or them getting together at all). Follow these probabilities back just a few generations and you get to anastronomical number. But that is not the point. Your existence wasn’t preordained. It wasn’t the goal. But it happened in spite of the astronomical odds against it.
OK. I can't believe it. You are really proposing again what I call "the infamous deck of cards fallacy". One of the worst and most arrogant wrong arguments that I have ever heard. It has been some time, maybe years, since I heard it the last time as a criticism of ID. It was rather frequent about 10 years ago, but then, apparently, even neo-darwinist must have realized how wrong and stupid it is. Not you, apparently. So, the fallacy goes as follows: We shuffle a deck of cards (let's say 52) and we draw all of them in random order (if we have shuffled well). What are the probablilities of that specific sequence to come out? Easy: this is a combinatorics problem too, permutations without repetition, and the asnwer is n! (n factorial). So, the answer is: 8.065818e+67 225 bits Well, that's not the 500 bits of Dembski's UPB, but I think it's enough. Almost everybody would agree with the folloing statement, taken from a web site:
To put this in perspective, the dinosaurs died out 65,000,000 years ago, and the age of the earth is just 4,500,000,000 years. Now suppose everybody in the world was to arrange packs of cards at the rate of one per second, it would take 600,000,000,000,000,000,000,000,000,000,000,000,000,000,000,000,000 years to get all the combinations! That's why you're VERY unlikely ever to shuffle a pack of cards the same way twice.
From: How many different ways can you put a pack of cards in order? http://www.murderousmaths.co.uk/cardperms.htm So, a really unlikely event. And here goes the infamous fallacy. It says: "See? the permutation you obtained is absoluitely unlikely, and yet you got exactly that. This is the demonstration that extremely unlikely events happen all the time!" And that is exactly your reasoning about me as an unlikely individual. Now, as narcissistic as I can be, I am not an unlikely individual. It's you who does not understand probability and specification (if you are sincere in what you say). I will assume you are sincere. So, how to explain it to you? There are many ways. I will try the simplest, referring again to my example. The thief and the safes. Beware: this is the explanation. Pay attention! So, our thief goes for the big safe (against all reason and common sense). OK, he is a neo-darwinist, and he has just read Joe Felsestein's arguments and your comment #850. He makes a first try, and he types a random 150 figures number. The safe does not open. What happened here? We have one event: the random generation of a 150 figures number. What is the probability of that event? It depends on how you define the probability. In all probability problems, you need a clear definition of what probability you are computing. So, if you define the problem as follows: "What is the probability of having exactly this result? ... (and here you must give the exact sequence for which you are computing the probability)" then the probability is 10^-150. But you have to define the result by the exact contingent information of the result you have already got. IOWs, what you are asking is the probability of a result that is what it is. That probability in one try is 1 (100%). Because all results are what they are. All results have a probability of 10^-150. That property is common to all the 10^150 results. Therefore, the probability of having one generic result whose probability is 10^-150 is 1, because we have 10^150 potential results with that property, and no one that does not have it. So, should we be suprised that we got one specific result, that is what it is? Not at all. That is the only possible result. The probability is 1. No miracle, of course. Not even any special luck. Just necessity (a probabiltiy of one is necessity). Now, let's say that our thief, at his first try, types exactly the sequence that opens the safe. Now we are defining the event not by some specific contingent sequence (we may have no idea at all of what the sequence is). We define it by something that it can do: IOWs a function. The sequence that opens the safe. The only sequence that can do that. What is the probability of getting that result? It is 10^-150! Really, this time. So, if our thief gets it at the first try, I will be really suspicious. The best explanation, by far, is that he already knew the solution (IOWs, design). See, the general concept is: what is a specification? the answer is: A specification is any objective rule that generates a binary partition in the search space. Now, both the definitions we have considered above do generate a binary partition in the search space. First definition: a result which is what it is, and is one of the 10^150 results that have an individual probability of 10^-150. Partition: Search space = 10^150 Target space = 10^150 The target space is the same as the search space. Probability of success = 1 (100%) Second definition: a result which can open the big safe. Partition: Search space = 10^150 Target space = 1 The target space is extremely small if compared to the search space. Probability of success = 10^-150 Therefore, the deck of cards fallacy is not only a fallacy: it is infamous, completely wrong and very, very silly and arrogant. It really makes me angry.gpuccio
April 11, 2018
April
04
Apr
11
11
2018
04:52 AM
4
04
52
AM
PDT
Allan Keith at #850: Two points: a) First point:
your question is obviously about the “combinatorial explosion”. So, I ask you a similar question in response.
You have not answered my question at #847. A "similar quesstion" is not an answer. You speak of "combinatorial explosion". I don't know if it is an "explosion", but it certainly is a very correct application of combinatorics, a well defined branch of mathematics. So, I ask again: What does he try? The big safe or the 150 smaller safes? What would you try? It's a simple question. Please, answer. The second point in next post.gpuccio
April 11, 2018
April
04
Apr
11
11
2018
03:37 AM
3
03
37
AM
PDT
bill cole- Allan Keith is "Acartia" over on TSZ. It has already admitted its purpose in these discussions is to be a pokey- do whatever it can to provoke people. Instigation is its name Just sayin'...ET
April 10, 2018
April
04
Apr
10
10
2018
08:20 PM
8
08
20
PM
PDT
Hi Allan
Calling it a strawman doesn’t make it so. The strawman is claiming that the combinatorial explosion disproves anything. The statistical assumptions used by those using this argument are wrong. Which makes the argument wrong. Don’t blame me if your assumptions are wrong.
The combinatorial explosion is a problem for identifying random change or trial and error as a cause of what is observed. When you talk about the cause of you being born we know what the cause is.:-) If you sit at a poker table and someone gets dealt 5 royal straight flushes in a row you would not assume a fair shuffled deck. The fact that he got those hands is 100% because it already happened the question, however, is the cause. The combinatorial explosion problem is not about backward probabilities it is about determining if random change can be the cause of the pattern you are seeing. When you see a functional protein sequence of 500 bits we can eliminate random change as a cause. This is one of the pieces of evidence that supports the design inference.bill cole
April 10, 2018
April
04
Apr
10
10
2018
07:17 PM
7
07
17
PM
PDT
Look, you are starting with existing humans who have the combinatorial explosion you need to account for in the first place. And then add in everything else I first posted in response to your straw man- that is the evidence it is a straw man. Just saying your "argument" is valid doesn't make it so.ET
April 10, 2018
April
04
Apr
10
10
2018
06:07 PM
6
06
07
PM
PDT
ET,
Your straw man has been exposed. Now run along and let us adults discuss science.
Calling it a strawman doesn’t make it so. The strawman is claiming that the combinatorial explosion disproves anything. The statistical assumptions used by those using this argument are wrong. Which makes the argument wrong. Don’t blame me if your assumptions are wrong.Allan Keith
April 10, 2018
April
04
Apr
10
10
2018
06:03 PM
6
06
03
PM
PDT
Your straw man has been exposed. Now run along and let us adults discuss science.ET
April 10, 2018
April
04
Apr
10
10
2018
05:44 PM
5
05
44
PM
PDT
ET,
The chances are high that a human will be born after a successful mating between a male and female human.
Thank you Captain Obvious.
The odds that any particular baby will have a unique genome is as close to 1 to 1 as you can get.
Duh! Your ability to comprehend is duly noted.Allan Keith
April 10, 2018
April
04
Apr
10
10
2018
05:28 PM
5
05
28
PM
PDT
Oh my, talk about a brain-dead argument. The chances are high that a human will be born after a successful mating between a male and female human. "What are the odds that you are going to be dealt a specific hand?" It is a certainty that I will be dealt a hand if I am in the game. The odds that any particular baby will have a unique genome is as close to 1 to 1 as you can get.ET
April 10, 2018
April
04
Apr
10
10
2018
04:35 PM
4
04
35
PM
PDT
Gpuccio, I apologize for not really following this thread, but your question is obviously about the “combinatorial explosion”. So, I ask you a similar question in response. You have a very unique and specific DNA sequence. What is the probability of this arising? Your father produced millions of sperm cells, your mother thousands of ova. To create you, the exact two would have had to get together. Add to this the probability of your parents getting together at the right time (or them getting together at all). Follow these probabilities back just a few generations and you get to anastronomical number. But that is not the point. Your existence wasn’t preordained. It wasn’t the goal. But it happened in spite of the astronomical odds against it.Allan Keith
April 10, 2018
April
04
Apr
10
10
2018
04:25 PM
4
04
25
PM
PDT
Allan Keith:
Maybe I am missing something.
A brain and a spine, at the very least. :razz:ET
April 10, 2018
April
04
Apr
10
10
2018
04:14 PM
4
04
14
PM
PDT
Allan Keith- Is that all you have? Really? You are lying, of course and slandering someone. Typical, but still pathetic. Wallow in your willful ignorance.ET
April 10, 2018
April
04
Apr
10
10
2018
04:12 PM
4
04
12
PM
PDT
Allan Keith: Waiting for Joe Felsestein, have you any position about what he says (quoted at #828) and my rebuttal at #828 and #831? I will ask the question again, to you, and in more detail. A thief enters a house, a big house. Inside, he knows that there are: a) One big safe, with a key that is some 150 figures number. b) 150 smaller safes, each of them with a key which is a one figure number: from 0 to 9. c) He knows that the same sum is in the big safe and in the 150 smaller safes put together. d) What does he try? The big safe or the 150 smaller safes? e) Of course, the gain in fitness is the same in both cases. You answer. Of course, maybe the thief is a proud fool, or maybe he is a neo-darwinist, and he chooses the big safe. But most thieves with a minimum of common sense would certainly go for the 150 smaller safes. A few reflections, waiting for your answer. We already know that Joe Felsestein apparently thinks that the two options are the same thing. If I have not misinterpreted what he says. A good thief, quick and concentrated and well organized could probably empty the 150 smaller safes in less than three hours, especially if he has a couple of accomplices to empty them while he opens them. Indeed, I think that about half a minute is needed to try the 10 digits, and most of the times he will find the right key in much less time. If he goes for the big safe, instead... OK, let's see. 10^150 combinations. Each of them 150 figures long. Let's say one minute a try, to be very generous. Reasonably, he could find the right key with half of the total possible number of tries. But we have faith in his moderate luck, so let's say that he can find the right combination after 1/10 of the possible tries. That leaves us with 10^149 tries. At one minute a try, that is... About 10^143 years. If the total time of our universe from the Big Bang to now is 1.5x10^10 years, then the time necessary to find the combination, with a little luck, is 10^133 times the whole lifetime of our universe. Some difference, with three hours! But of course neo-darwinists understand very well that 150 events with a complexity of 3.3 bits each (the complexity of a decimal figure) are the same thing as one event with a complexity of 500 bits. Joe Felsestein seems to defend this position, and nobody on his side has apparently disagreed. What do you say? I think this is a very important point. I would like to hear statements about that.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
03:48 PM
3
03
48
PM
PDT
ET,
Already done, Allan. And it was posted on TSZ- and guess what? They all choked on it.
Maybe I am missing something. Both of those links were to a site who’s owner is a well known jerk who does nothing but insult and swear at anyone who disagrees with him. A well known homophobe who frequently calls his opponents “faggots”, “assmunchers” and other more offensive epithets. Do yo have links to any more reputable sites? I would be interested to read those.Allan Keith
April 10, 2018
April
04
Apr
10
10
2018
03:40 PM
3
03
40
PM
PDT
Already done, Allan. And it was posted on TSZ- and guess what? They all choked on it. How to test and falsify ID Definitive evidence that ATP synthase was intelligently designed And that is more than evolutionism has. There isn't anything published that supports evolutionism. There isn't a methodology to test the claim that natural selection produced vision systems, for example.ET
April 10, 2018
April
04
Apr
10
10
2018
03:20 PM
3
03
20
PM
PDT
OMagain:
It always amuses me to ask “which one?” when IDists proclaim that the bacterial flagellum was designed.
And it always amuses us when you try to defend the claim tat any one of them evolved via natural selection and/ or drift. Your entire position is amusing because it always comes back to flailing away at ID with your ignorance. Even if you ever find a valid fault with ID that will never help you find support for your position's claims.ET
April 10, 2018
April
04
Apr
10
10
2018
03:17 PM
3
03
17
PM
PDT
ET,
You want to watch TSZ implode? We need to post the way to test ID, along with the positive criteria. Who do I talk to?
God? But seriously, we would love to see ID start to do this. When are you going to start? Feel free to start with ATP synthase. How do you test that ID created it? Feel free to publish this in any of the high end peer reviewed journals. If you can’t do that, feel free to publish your research in BioComplexity. Or, if you can’t get it published there, just post it here.Allan Keith
April 10, 2018
April
04
Apr
10
10
2018
03:12 PM
3
03
12
PM
PDT
You want to watch TSZ implode? We need to post the way to test ID, along with the positive criteria. Who do I talk to?ET
April 10, 2018
April
04
Apr
10
10
2018
02:29 PM
2
02
29
PM
PDT
DNA_Jock at TSZ: April 10, 2018 at 8:41 pm Please, read my answer at #834.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:56 PM
12
12
56
PM
PDT
OMagain:
It’s kind of the point actually. Multiple ways of doing similar things is the ladder.
Not at all. Gradual stepwise naturally selectable configurations of a sequence, linked by simple variation and which show a constant increase of the selectable function: that's a ladder. I don't know where you have left your logic.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:54 PM
12
12
54
PM
PDT
Corneel at TSZ: Thank you for signaling the typo. I have just deleted the "y"! :) I am a bad monkey. No Shakespeare from me, certainly! :)gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:47 PM
12
12
47
PM
PDT
DNA_Jock at TSZ: <blockquote cite<It’s not like we haven’t been over this before. Sigh. See my comment #837.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:43 PM
12
12
43
PM
PDT
dazz:
Here we go again with the caricature of a straw man, sprinkled with tons of texas sharp shooting
I am writing a full OP about the TSS fallacy and other arguments from DNA_Jock. Please, have a little patience.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:41 PM
12
12
41
PM
PDT
Corneel at TSZ:
Just a warning.
Consider me warned, but not impressed. Yes, the variance is different in different gorups, but there can be many different explanations for that. One of them could be that the groups have very different mumerosity (not my fault, it depends on the number of sequenced data in each group). You should also consider that the analysis regards the whole human proteome, about 20000 proteins. It's a rather good population size. I really see no problems here.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:39 PM
12
12
39
PM
PDT
Corneel at TSZ: You can also look at my comments #828 and #831 for an interesting follow-up to my discussion with Joe Felsestein.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:33 PM
12
12
33
PM
PDT
Corneel at TSZ:
So those naughty E3 ligases are promiscuous as well, and they may acquire novel targets simply by having their expression changed to another cellular compartment or different timing. Regulation which may be changed by single nucleotide changes in the regulatory domain of those genes. Of course, ensuing evolution towards greater affinity can occur in small evolutionary steps.
This strange comment should be justified by the following passage in the paper you quote.
3. Significant degrees of redundancy and multiplicity. Any particular substrate may be targeted by multiple E3 ligases at different sites, and a single E3 ligase may target multiple substrates under different conditions or in different cellular compartments. This drives a huge diversity in spatial and temporal control of ubiquitylation (reviewed by ref. [61]). Cellular context is an important consideration, as substrate–ligase pairs identified by biochemical methods may not be expressed or interact in the same sub-cellular compartment.
Well, I have see many non sequitur, but this is one of the best. So, we have 600+ E3 ligases which are completely different one from the other at sequence level (except for the shraed, small domains, as explained). Now we learn that: "Any particular substrate may be targeted by multiple E3 ligases at different sites," and: "a single E3 ligase may target multiple substrates under different conditions or in different cellular compartments" My original statement was: "Not so in the case of the specificity of E3 ligases. That specificity is about recognizing completely different target proteins, and their appropriate state." Emphasis added. So, a single E3 ligase can target more than one protein. But that was already clear, because we have 600+ E3 ligases and thousands of targets. See the OP:
2 E1 proteins – 40 E2 proteins – 600+ E3 proteins + thousands of specific substrates IOWs, each of hundreds of different complex proteins recognizes its specific substrates, and marks them with a shared symbolic code based on uniquitin and its many possible chains. And the result of that process is that proteins are destined to degradation by the proteasome or other mechanisms, and that protein interactions and protein signaling are regulated and made possible, and that practically all cellular functions are allowed to flow correctly and smoothly.
Emphasis added. That perfectly corresponds to: "a single E3 ligase may target multiple substrates under different conditions or in different cellular compartments". But we also learn that: "Any particular substrate may be targeted by multiple E3 ligases at different sites," (emphasis added) So, it's not so much redundancy, as multiplicity. Different E3 ligases may target a same protein, but at different sites. Specificity, again. You say that they are "promiscuous". But the authors have different conclusions, and I agree fully with them: "This drives a huge diversity in spatial and temporal control of ubiquitylation (reviewed by ref. [61]). Cellular context is an important consideration, as substrate–ligase pairs identified by biochemical methods may not be expressed or interact in the same sub-cellular compartment." (Emphasis mine) IOWs, there is a huge diversity of strict control: IOWs, huge functional complexity. Promiscuous? Do you know how that is called? It's called "cross-talk". A term that we have found consistently in the scientific literature about the ubiquitin system. It's semiosis and complexity, at the highest level. How you can ifer from these interesting observations that: "Of course, ensuing evolution towards greater affinity can occur in small evolutionary steps." is really a mystery, to me.
You did not offer protein domains as an example, gpuccio. You offered proteins. The function of a protein relies for a great deal on the specific combination of domains it has.
Yes. And so? What do you mean. I stick to all that I have said about this issue.
Did you read Joe Felsensteins criticism in the beginning of the thread?
Yes, of course. And I have answered it in great detail. Did you read my answer to him at #716?gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:31 PM
12
12
31
PM
PDT
1 2 3 4 5 6 32

Leave a Reply