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
a deubiquitinating enzyme complex regulates the mitotic spindle assembly factor NuMA. BRISC binds and deubiquitinates the spindle assembly factor NuMA
Spindle assembly at a BRISC pace Compared with a control cell (left), microtubules (green) form a multipolar spindle in a cell (right) lacking the BRISC component ABRO1 (red). DNA is labeled blue. Yan et al. describe how a deubiquitinating enzyme complex regulates the mitotic spindle assembly factor NuMA. BRCC36 is a deubiquitinating enzyme that preferentially cleaves lysine 63–linked polyubiquitin chains. As part of the Rap80 complex, BRCC36 regulates the repair of DNA double-strand breaks, but the enzyme can also assemble into a distinct complex, known as BRISC. Yan et al. found that knocking down BRCC36, or a subunit unique to the BRISC complex called ABRO1, caused mitotick cells to assemble multipolar spindles that frequently aligned and segregated chromosomes incorrectly. These defects could not be rescued by a catalytically inactive version of BRCC36. In wild-type mitotic cells, BRISC accumulated at the spindle poles, bound to the minus ends of stable, kinetochore-attached microtubules. Early in mitosis, BRISC also localized near the kinetochores themselves, where it promoted the chromosome-dependent nucleation of spindle microtubules. Yan et al. discovered that BRISC binds and deubiquitinates the spindle assembly factor NuMA, which captures and focuses microtubules at spindle poles. In the absence of BRISC, ubiquitinated NuMA showed an increased association with both importin-? and dynein, which regulate the protein’s function. BRISC therefore promotes bipolar spindle assembly by deubiquitinating NuMA. Senior author Genze Shao says that BRISC may have other mitotic substrates as well. Because some BRISC-deficient cells progress through mitosis, despite their disorganized spindles, he is particularly interested in whether the deubiquitinase can regulate the spindle assembly checkpoint. •
• Yan, K., et al0 . 2015. J. Cell Biol. doi:10.1083/jcb.201503039 • Ben Short
Dionisio
February 19, 2018
February
02
Feb
19
19
2018
07:34 PM
7
07
34
PM
PDT
The cell division cycle is driven by a collection of enzymes that coordinate DNA duplication and separation, ensuring that genomic information is faithfully and perpetually maintained. The activity of the effector proteins that perform and coordinate these biological processes oscillates by regulated expression and/or posttranslational modifications. Ubiquitylation is a cardinal cellular modification and is long known for driving cell cycle transitions. In this review, we emphasize emerging concepts of how ubiquitylation brings the necessary dynamicity and plasticity that underlie the processes of DNA replication and mitosis. New studies, often focusing on the regulation of chromosomal proteins like DNA polymerases or kinetochore kinases, are demonstrating that ubiquitylation is a versatile modification that can be used to fine-tune these cell cycle events, frequently through processes that do not involve proteasomal degradation. Understanding how the increasing variety of identified ubiquitin signals are transduced will allow us to develop a deeper mechanistic perception of how the multiple factors come together to faithfully propagate genomic information. Here, we discuss these and additional conceptual challenges that are currently under study toward understanding how ubiquitin governs cell cycle regulation.
Gilberto, Samuel & Peter, Matthias. (2017). Dynamic ubiquitin signaling in cell cycle regulation. The Journal of Cell Biology. 216. jcb.201703170. 10.1083/jcb.201703170. http://jcb.rupress.org/content/216/8/2259.full-text.pdf
Dionisio
February 19, 2018
February
02
Feb
19
19
2018
07:16 PM
7
07
16
PM
PDT
This thread has too much bad news for the 'modern synthesis' and the 'third way' clubs. :)Dionisio
February 19, 2018
February
02
Feb
19
19
2018
07:12 PM
7
07
12
PM
PDT
gpuccio @17: That figure 1 legend has material to fill post-doc course textbooks.Dionisio
February 19, 2018
February
02
Feb
19
19
2018
07:04 PM
7
07
04
PM
PDT
Mitosis ensures equal segregation of the genome and is controlled by a variety of ubiquitylation signals on substrate proteins. However, it remains unexplored how the versatile ubiquitin code is read out during mitotic progression. Here, we identify the ubiquitin receptor protein UBASH3B as an important regulator of mitosis. UBASH3B interacts with ubiquitylated Aurora B, one of the main kinases regulating chromosome segregation, and controls its subcellular localization but not protein levels. UBASH3B is a limiting factor in this pathway and is sufficient to localize Aurora B to microtubules prior to anaphase. Importantly, targeting Aurora B to microtubules by UBASH3B is necessary for the timing and fidelity of chromosome segregation in human cells. Our findings uncover an important mechanism defining how ubiquitin attachment to a substrate protein is decoded during mitosis.
Ubiquitin Receptor Protein UBASH3B Drives Aurora B Recruitment to Mitotic Microtubules. Krupina, Ksenia & Kleiss, Charlotte & Metzger, Thibaud & Fournane, Sadek & Schmucker, Stephane & Hofmann, Kay & Fischer, Benoit & Paul, Nicodeme & Porter, Iain & Raffelsberger, Wolfgang & Poch, Olivier & Reese Swedlow, Jason & Brino, Laurent & Sumara, Izabela. (2016). Developmental Cell. 36. 63-78. 10.1016/j.devcel.2015.12.017. http://www.cell.com/developmental-cell/pdf/S1534-5807(15)00826-6.pdf
Dionisio
February 19, 2018
February
02
Feb
19
19
2018
06:58 PM
6
06
58
PM
PDT
gpuccio @16:
There are a lot of highly amazing things in biology
That's an understatement. :)Dionisio
February 19, 2018
February
02
Feb
19
19
2018
06:54 PM
6
06
54
PM
PDT
And, of course, deubiquitinating enzymes have a key role in regulating DNA repair. See here: Fine-tuning the ubiquitin code at DNA double-strand breaks: deubiquitinating enzymes at work https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4561801/ If you love simplicity, just look at Figure 1! And carefully read the Figure legend... :)gpuccio
February 19, 2018
February
02
Feb
19
19
2018
08:38 AM
8
08
38
AM
PDT
butifnot: There are a lot of highly amazing things in biology. But we have to dig deep into them, to really understand and appreciate their complexity and value. As in many other contexts, the devil is in the details! :)gpuccio
February 19, 2018
February
02
Feb
19
19
2018
05:18 AM
5
05
18
AM
PDT
This is the most amazing thing I have ever heard.butifnot
February 19, 2018
February
02
Feb
19
19
2018
05:14 AM
5
05
14
AM
PDT
In the OP I have barely mentioned the role of ubiquitination in DNA repair. This is interestng, because ubiquitination acts mainly at the level of histones. We know that Post Translational Modifications (PTMs) of histones are in themselves a code, the histone code. This code is, too, highly symbolic, and very complex indeed. See, for example, Wikipedia: https://en.wikipedia.org/wiki/Histone_code However, the histone code is mainly written by simpler PTMs, especially methylation (mono, di and tri methylation) and acetylation. The role of ubiquitin here is minor. But in the case of DNA damage, especially Double-Strand Breaks (DSB), ubiquitin becomes the absolute protagonist, and directs the repair process in an extremely complex and detailed way. See, for example, this paper: Writers, Readers, and Erasers of Histone Ubiquitylation in DNA Double-Strand Break Repair https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4923129/ The main role seems to be played by two E3 ligases, RNF8 and RNF 168, but a lot of other proteins are involved. So, this is another example of two complex symbolic codes (histone code and ubiquitin code) strongly interacting, in a highly dynamic and articulated pattern.gpuccio
February 19, 2018
February
02
Feb
19
19
2018
04:31 AM
4
04
31
AM
PDT
I have added to the OP a beautiful animation of the ubiquitin-proteasome system in degrading damaged proteins.gpuccio
February 18, 2018
February
02
Feb
18
18
2018
10:07 PM
10
10
07
PM
PDT
About branched ubiquitin chains and, again, NF-kB regulation: The K48-K63 Branched Ubiquitin Chain Regulates NF-kB Signaling. http://www.cell.com/molecular-cell/fulltext/S1097-2765(16)30563-9 (Public access) Abstract:
Polyubiquitin chains of different topologies regulate diverse cellular processes. K48- and K63-linked chains, the two most abundant chain types, regulate proteolytic and signaling pathways, respectively. Although recent studies reported important roles for heterogeneous chains, the functions of branched ubiquitin chains remain unclear. Here, we show that the ubiquitin chain branched at K48 and K63 regulates nuclear factor ?B (NF-?B) signaling. A mass-spectrometry-based quantification strategy revealed that K48-K63 branched ubiquitin linkages are abundant in cells. In response to interleukin-1?, the E3 ubiquitin ligase HUWE1 generates K48 branches on K63 chains formed by TRAF6, yielding K48-K63 branched chains. The K48-K63 branched linkage permits recognition by TAB2 but protects K63 linkages from CYLD-mediated deubiquitylation, thereby amplifying NF-?B signals. These results reveal a previously unappreciated cooperation between K48 and K63 linkages that generates a unique coding signal: ubiquitin chain branching differentially controls readout of the ubiquitin code by specific reader and eraser proteins to activate NF-?B signaling.
gpuccio
February 18, 2018
February
02
Feb
18
18
2018
09:57 PM
9
09
57
PM
PDT
This is a funny video. She does not use Lego, but something like that... https://www.youtube.com/watch?v=miZYmuDKO2s About histone ubiquitination, which I have not touched in the OP, here is an interesting paper: "Histone Ubiquitination and Deubiquitination in Transcription, DNA Damage Response, and Cancer" https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3355875/ (Public access) Both monoubiquitination and polyubiquitination seem to happen at histone level.gpuccio
February 18, 2018
February
02
Feb
18
18
2018
01:13 PM
1
01
13
PM
PDT
I think that the title of the following 2016 paper about the ubiquitin system could be of some interest: "Design Principles Involving Protein Disorder Facilitate Specific Substrate Selection and Degradation by the Ubiquitin-Proteasome System" https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4807260/ (Emphasis mine. Public access.) The Abstract is very interesting, too:
The ubiquitin-proteasome system (UPS) regulates diverse cellular pathways by the timely removal (or processing) of proteins. Here we review the role of structural disorder and conformational flexibility in the different aspects of degradation. First, we discuss post-translational modifications within disordered regions that regulate E3 ligase localization, conformation, and enzymatic activity, and also the role of flexible linkers in mediating ubiquitin transfer and reaction processivity. Next we review well studied substrates and discuss that substrate elements (degrons) recognized by E3 ligases are highly disordered: short linear motifs recognized by many E3s constitute an important class of degrons, and these are almost always present in disordered regions. Substrate lysines targeted for ubiquitination are also often located in neighboring regions of the E3 docking motifs and are therefore part of the disordered segment. Finally, biochemical experiments and predictions show that initiation of degradation at the 26S proteasome requires a partially unfolded region to facilitate substrate entry into the proteasomal core.
(Emphasis mine) The theme of intrinsically disordered regions in proteins is becoming ever more relevant. See also here: "Classification of Intrinsically Disordered Regions and Proteins" https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4095912/ Public access. I quote just the title of the first section: 1.1. Uncharacterized Protein Segments Are a Source of Functional Noveltygpuccio
February 18, 2018
February
02
Feb
18
18
2018
12:45 PM
12
12
45
PM
PDT
Dionisio: That will perhaps be pleasant news to some folks, but believe me, you will be missed by many others, including me. I am happy that your project is proceeding. However, I hope that you can still find some time to be with us, maybe with less frequency and volume, but with your usual ingenuity! :)gpuccio
February 18, 2018
February
02
Feb
18
18
2018
07:03 AM
7
07
03
AM
PDT
gpuccio, Excellent OP, as usual. Thanks. Also very interesting discussion with DATCG, who writes insightful comments and pointed to an interesting biology video series on the same fascinating topic of the OP. Here's a link to a full text PDF copy of one of the papers you referenced in the OP: https://www.researchgate.net/profile/Lakshminarayan_Iyer/publication/221848141_Structure_and_Evolution_of_Ubiquitin_and_Ubiquitin-Related_Domains/links/57436c4a08aea45ee84d1061/Structure-and-Evolution-of-Ubiquitin-and-Ubiquitin-Related-Domains.pdf Note the 21 citations to the given paper. PS, my current activities are keeping me from commenting lately. Perhaps that's pleasant news to some folks out there who were annoyed by my posts and even kept statistical track of their frequency and volume, but never dared to address them seriously. What else is new? The project I've been working on has proceeded to another phase that requires more attention to difficult for me issues.Dionisio
February 18, 2018
February
02
Feb
18
18
2018
06:51 AM
6
06
51
AM
PDT
DATCG: The second video is very interesting too. Starting at 1:33, the structure of Cal1 E3 ligases (of which the one shown at Fig. 7 in the OP is a very good example) seems to be a wonderful example of modular design. 70 F-boxes. 40 SOCS boxes. 250 different assemblies. Really amazing!gpuccio
February 18, 2018
February
02
Feb
18
18
2018
06:41 AM
6
06
41
AM
PDT
DATCG: At timepoint 23:25 of the first video there is a brief explanation of the role of phosphorylation in regulating ubiquitination. I have given an example of that in the OP, when I speak of the regulation of NF-kB, where phosphorylation provides the ubiquitination activating signal, both in the canonical and non canonical pathway, acting on the substrate. As shown in the video, phosphorylarion can act both on the substrate and on the E3 ligase, and finally it can also have an inhibitory role. This is fascinating, because we have a simpler semiotic system (phosphorylation) which controls a much more complex semiotic system (ubiquitination) to regulate the working of an effector system (NF-kB signaling) which is, itself, highly semiotic.gpuccio
February 18, 2018
February
02
Feb
18
18
2018
04:22 AM
4
04
22
AM
PDT
DATCG: At timepoint 2:39 of the first video there is already a very important point which deserves discussion: why proteins, and especially regulator proteins, need to be unstable so that adjustments in their steady state can be achieved quickly. It means that biological systems invest a lot of resources to achieve flexibility and quick (and very intelligent) adaptation to changing conditions. I suppose that can be achieved because one of life's features is to depend so critically on far from equilibrium processes. Food for thought.gpuccio
February 18, 2018
February
02
Feb
18
18
2018
12:19 AM
12
12
19
AM
PDT
DATCG: Thank you for the link to the video. I have added it (alll three parts) at the end of the OP. I have not yet had the time to see them all, but I will, and I will comment on the points I find most interesting! :) Yes, codes are of supreme importance in cellular life, and they do scream design. You say: "Conditional processing is a feature here I think as well as specificity." That's exactly the point! This ubiquitin code is a perfect implementation of conditional processing, an equivalent of complex "if - else" structures. Using an universal tag with internal configurable switches is really a brilliant solution for that. And it also reminds me of Lego construction toys! :)gpuccio
February 17, 2018
February
02
Feb
17
17
2018
10:38 PM
10
10
38
PM
PDT
Looking forward to it. Thanks again for your interesting post. And yes, very much like this approach. Hope others join in. Nothing like peering through signals, interpretations and post-translations and meta layers of information code on top of code. Conditional processing is a feature here I think as well as specificity. Agree with you. Not judging Arthur. If my speculation is correct, then I'm defending him. And hope he may show up in the future. But onward :) To the Ubiquitin Code. And hope it's OK to add a video overview in 3 parts for those who might like to see some animation along with explanations. 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... https://www.youtube.com/watch?v=ILdEOXCfgUc There's more of course, but thought he added some good insights.DATCG
February 17, 2018
February
02
Feb
17
17
2018
05:18 PM
5
05
18
PM
PDT
DATCG: Thank you for opening the discussion! :) I understand that the OP is rather long and touches a lot of stuff. I hope that you and others who are interested in this kind of approach can give a very good contribution to a discussion about this fascinating issue. Of course, UB is specially invited to comment: he is the master of the semiosis arguments, and I am sure that he will be interested in the things discussed here. :) Like you, I have been a little disappointed of the apparent disappearance of Arthur Hunt from the spliceosome discussion. He could certainly have given a great contribution. However, not knowing his reasons, I cannot certainly judge.gpuccio
February 17, 2018
February
02
Feb
17
17
2018
03:43 PM
3
03
43
PM
PDT
Oooooooooooooohhhh more fun :) I need time to read later and look forward to another good discussion. I made a quick comment on Spliceosome post by you and scientist Arthur Hunt's disappearance from the discussion a moment ago. Scientist Arthur Hunt: https://www.researchgate.net/profile/Arthur_Hunt Thought he would contribute to a good debate and discussion points in opposition to you Gpuccio on the Spliceosome, it's evolution, Design or not. But he seems to have disappeared without any rebuttal to your detailed responses... Spliceosome - Defy non-Design Explanations .DATCG
February 17, 2018
February
02
Feb
17
17
2018
03:31 PM
3
03
31
PM
PDT
1 30 31 32

Leave a Reply