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
Corneel at TSZ: So why would the protein coding genes not be representative of all evolutionary transmitted information? I trust that you have a scientific and empirical argument for doubting this. Yes. By far the most important argument is that I don't believe that the structure of the complex nervous system in humans, in particular the brain, and its exclusive new potentialities, can derive from a very small change in protein coding genes. For that, even the changes in regulatory non coding sequences seem to be insufficient. I do believe that it is transmitted information, but I don't know where it is and how it is transmitted. That's not so strange. A lot of obviously transmitted information in living organisms is completely elusive, at present: for example, complex behaviours in some types of orgnaisms.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:08 PM
12
12
08
PM
PDT
Intelligent Design is about the DESIGN. ID is not about the designer(s) because we don't even ask about that until after we have determined intelligent design exists. Yes, the existence of intelligent design says there was an intelligent designer. But we only have the DESIGN to study.ET
April 10, 2018
April
04
Apr
10
10
2018
09:14 AM
9
09
14
AM
PDT
Joe Felsenstein at TSZ: Just to make it simple for all. Please, read my comment #823. It's short and simple. OK, I paste it again here.
Of course 500 bits refers to an exponential measure for one object (or set of objects, if we can show that the set is IC) implementing one specific function. It’s the implemention of one function which has a complexity of 500 bits: IOWs, it cannot be implemented with a simpler configuration. Independent individual bits cannot be summed. Bits are an exponential measure. 500 bits means: a specific sequence of 500 binary values, or of 150 specific decimal values, or of 115 specific AAs (base 20) that is necessary to implement one specific function. It’s like having a number like this: 6394672650104367823952223904758… 150 figures long, which is the unique key to an electronic safe, and trying to divine it by RV and NS: a) RV: we just try any sequence of 150 figures b) NS: after seeing it does not work, we change a figure at a time, and we test if there is any increase in the (non) function. This is what the neo-darwinian model amounts to, as far as complex functional proteins are involved. Has Joe Felsestein any comment?
OK, now a very simple question. Is finding the 150 figures key the same as finding 150 different one figure keys to 150 different safes? In both scenarios, the "fitness" of the thief increases, I suppose. Answer, please.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
08:44 AM
8
08
44
AM
PDT
ET: "I tried to warn you." That's true! :)gpuccio
April 10, 2018
April
04
Apr
10
10
2018
08:36 AM
8
08
36
AM
PDT
Are you kidding? Have you lost your mind?
I tried to warn you.ET
April 10, 2018
April
04
Apr
10
10
2018
08:32 AM
8
08
32
AM
PDT
Joe Felsenstein at TSZ: April 10, 2018 at 12:32 am Are you kidding? Have you lost your mind?
The 500 bits criterion, which originated with Dembski, was gpuccio’s criterion for “complex”, as I demonstrated in clear quotes from gpuccio in my previous comment.
It's of course my criterion for complex functional information: the information linked to the implementation of one explicitly defined function. IOWs, if a function can be implemented by an object, or by an IC system, and it requires at least 500 specific bits to be implemented by that object, it is complex. As I have always said. See also one of my first OPs here: Functional information defined https://uncommondescent.com/intelligent-design/functional-information-defined/
That counts up changes anywhere in the genome, as long as they contribute to the fitness, and it counts up whatever successive changes occur.
Again, are you kidding? So, if you have 500 different mutations of 1 AA in different proteins, each of them contributing in completely different and independent ways to fitness, you believe that you have 500 bits of complex functional information? Are you really saying that? I cannot believe it! Each of those mutations is independent, and has an independent and different functional effect. Each of them contributes to fitness, therefore is selectable. None of them contributes to the same function as the others, even if all of them contribute, independently, to fitness (which is of course a meta-function, ot which many different functions contribute). Do you understand why we measure functional information (yes, the same functional information that you recognized as a true and important concept) in bits? It's because it is -log2 of the probability of the event. Do you understand? Bits are exponential. It is rather easy to have one random AA change which is specific to one function (there is not function, or increase of the function, without it). It is more difficult to have two random AA changes that are specific to one function (there is not function, or increase of the function, without both of them). It is empirically impossible to have 150 random AA changes that are specific to one function (there is not function, or increase of the function, without all of them). The sum of 150 simple mutations, each of which gives inependently an increse of "fitness", is building no complex function. The idea of neo-darwinism is that a complex function (like ATP synthase) should come into existence through hundreds of specific mutations in the same structure which in the end build the function as we observe it today. And each of those mutations should increase fitness, and therefore be naturally selectable. Now, either each mutation is naturally selectable because the final function already exists: IOWs, ATP synthase appears as a simple mutation (5 AAs top) in something unrelated that already existed, and the humdreds of specific AAs that follow just "optimize" that simple initial function Or The final function (ATP synthase) just appears when hundreds of specific AAs are in place, but for some strange reason there is a ladder of simple mutations, each of them increasing fitness for different and inscrutable reasons, which for some even stranger reason just builds the exact complex sequence that will, one day, provide a completely different function, ATP synthase. You choose which of the two is less empirically impossible.
Now, in both gpuccio’s and your comments, the requirement is added that all this occur in one protein, in one change, and that it be “new and original function”.
It is not added. It was there from the beginning. Just read any single discussion I have had here in the last ten years. Or just my linked OP about functional complexity. Or anythin from me about the issue.
That was not a part of the 500-bit criterion that gpuccio firmly declares to be the foundation of ID.
It was, of course. It's not my fault if you don't understand ID theory. Not at all.
There was supposed to be some reason why a 500 bit increase in functional information was not attainable by natural selection. Without any requirement that it involve “new and original function”.
I have explained what new and original mean, and why they are part of the definition of functional information from the beginning. See also comment #716, to you.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
08:30 AM
8
08
30
AM
PDT
Entropy at TSZ: April 10, 2018 at 12:31 am You seem to have lost any reasonable attitude, not for the first time I must say.
A few curious things, 1. The ubiquitin system, and the sequence of ubiquitins themselves, are described as very conserved. 2. The existence of distant homologs for all of the proteins involved in the ubiquitin systems is acknowledged, thus contradicting 1. 3. The distant homologs are claimed to “add to the complexity of the system” 4. But gpuccio insists that gene copies do not add functional information/complexity. Thus contradicting 3. 5. This guy rejects enzyme promiscuity as a “conceptual reason” for the existence of “ladders” towards “complex protein function” on the basis of the similarity of the substrates recognized by those enzymes. 6. Yet he’s impressed by the way the ubiquitin system works, even though it involves proteins belonging to larger protein families, similar substrates, similar reactions, and similar actions.
Curious things? 1. Ubiquitin, the molecule, is extremely conserved. The other components are more or less conserved, in different ways. For example, the set of 600+ E3 ligases shows very different conservation history, and that is an explicit point in my OP, and especially in Fig. 5. 2. I have ackowledged the existnce of distant homologues for ubiquitin itself, ubiquitin like proteins, and Probably some other components. E3 ligases, for example, are not present in prokaryotes at all (there is only an example of a RING domain, but not of an E3 ligase, as already discussed. See the OP section: "Evolution of the ubiquitin system?" and my comment 758 to Corneel. How these facts "contradict" the high conservation of ubiquitin in eukaryotes, is really a mystery. 3. The phrase you quote is, apparently, from the OP: "A number of ubiquitin like proteins add to the complexity of the system." Ubiquitin like proteins are not "distant homologs". They are more variant molecules, often rather unrekated to the ubiquitin molecule, which have a distinct role in the system, separated from the role of ubiquitin. That's why they "add to the complexity". Of course they do. And, like ubiquitin, some Ubl have distant homologues in prokaryotes, as you can check in the abstract of the paper quoted in the section "Evolution of the ubiquitin system?" 4. They are not gene copies. They are different and specialized molevule, each with its specialized system. not contradiction. 5. Yes. And also on the base of the extreme similarity of the proteins themselves, which are usually part of a same protein family, or even sub-family. 6. I am amazed at many things. The greatest of all, in the ubiquitin system, is the huge group of the E3 ligases, as said many times. E3 ligases share some basic domains for the function of ubiquitin transfer, domains which are however of a few different basic types, but they are completely different one from the other for the main part of the molecule, which has the role of recognizing the correct target. But why should I explain it again? I can just quote my OP, where the issue is clearly explained:
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: 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) 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. 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.
Other curious things?gpuccio
April 10, 2018
April
04
Apr
10
10
2018
07:57 AM
7
07
57
AM
PDT
corny the quote-mining fool just cannot help itself:
He really seems incapable of seeing the delicious irony of him proudly proclaiming that they establish their “scientific methodology” AFTER they have decided that organisms are designed.
That is your opinion and it is incorrect. ID's scientific methodology is geared toward distinguishing intelligent design from nature. That much has been spelled out in every piece of pro-ID literature. And when compared to your position, which only has, "anything but ID" any objective person can see which one is science and which one is a joke.ET
April 10, 2018
April
04
Apr
10
10
2018
06:23 AM
6
06
23
AM
PDT
My favorite part for identifying ATP synthase as intelligently designed is the external connection between the two functional subunits that has nothing to do with the functionality of either subunit but without which ATP synthase would not exist: he architecture and subunit composition of ATP synthase It holds the two subunits just the right distance apart so that together they form ATP synthase. Without it the cap would just float freely around the dead cellET
April 10, 2018
April
04
Apr
10
10
2018
06:14 AM
6
06
14
AM
PDT
ET (and Joe Felsestein at TSZ): The alpha and beta chains of ATP synthase remain a good example.gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:16 AM
12
12
16
AM
PDT
ET (and Joe Felsestein at TSZ): Of course 500 bits refers to an exponential measure for one object (or set of objects, if we can show that the set is IC) implementing one specific function. It's the implemention of one function which has a complexity of 500 bits: IOWs, it cannot be implemented with a simpler configuration. Independent individual bits cannot be summed. Bits are an exponential measure. 500 bits means: a specific sequence of 500 binary values, or of 150 specific decimal values, or of 115 specific AAs (base 20) that is necessary to implement one specific function. It's like having a number like this: 6394672650104367823952223904758... 150 figures long, which is the unique key to an electronic safe, and trying to divine it by RV and NS: a) RV: we just try any sequence of 150 figures b) NS: after seeing it does not work, we change a figure at a time, and we test if there is any increase in the (non) function. This is what the neo-darwinian model amounts to, as far as complex functional proteins are involved. Has Joe Felsestein any comment?gpuccio
April 10, 2018
April
04
Apr
10
10
2018
12:14 AM
12
12
14
AM
PDT
Joe F's misconception of CSI summed up:
2. That counts up changes anywhere in the genome, as long as they contribute to the fitness, and it counts up whatever successive changes occur.
No Joe. No where does Dembski ever make that claim. Reading Dembski and Meyer they both make it clear they are talking about producing the 500 bits in one sequence.ET
April 9, 2018
April
04
Apr
9
09
2018
04:58 PM
4
04
58
PM
PDT
dazz at TSZ: This is an interesting passage from the paper quoted at #803:
Systematic studies on the evolutionary origin of orphan genes in primates (Toll-Riera et al. 2009) and the plant Arabidopsis thaliana (Donoghue et al. 2011) indicate that gene duplication and exaptation from transposable elements (TEs) are the major forces driving the emergence of orphan genes. Another study investigating the emergence of new Drosophila genes (not restricted to orphan genes) corroborated the dominant role of gene duplication but also suggested that surprisingly many genes (~12%) seem to have originated de novo, that is, from previously noncoding sequences or RNA coding sequences (CDS) (Zhou et al. 2008).
gpuccio
April 9, 2018
April
04
Apr
9
09
2018
01:46 PM
1
01
46
PM
PDT
dazz at TSZ:
Well, that’s appalling. The way I see it you’re disagreeing with yourself since I’m drawing conclusions from your claims
I am disagreeing with how you draw conclusions from my claims.
Unfortunately, no new, original functionally specified complexity of a gazillion bits of information was generated in that microevolutionary event as far as I can tell.
What micorevolutionary event? The generation of a new functional ORF in a non coding sequence is not a microevolutionary event. The final step that relaeses it as a transcirbed sequence is just the final step, but it would be useless if the sequence did not code for a functional protein. The whole process is not microevolutionary.
You’re obsessed with neo-darwinism, but evolutionary changes don’t need to be selectable at every single step. Neutral theory and all that, no “design” in sight
If you appeal to neutral theory, the probabilistic barriers remain the same. You have to accept the limitations of RV. If you appeal to NS, you have to accept the limitations of NS. If you put both together, you have the limitations of the combined algorithm: only simple evolutionary events are allowed.
We seem to be in disagreement as to what gradual means. It may develop in a month, but there is a continuum of small gradual changes, with lots of IC generation and apparently no issues with lack of protein functionality due to islands of function. Maybe most of those transitions you consider problematic are essentially developmental and you’re grossly overstating the importance of “new” protein function?
I don't think so.
You’re the one who’s confused. If, as I argue follows from your premises and claims, saltation is inevitable, the existence of intermediates would readily falsify your claims. It’s not rocket science
You are right. I had misunderstodd your statement. I apologize. You will find the correct answer at #815.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
01:40 PM
1
01
40
PM
PDT
gpuccio- It has nothing to do with any debate with Dembski. CSI is Dembski's construct and it is important to how your opponents view that construct in order to know what they are talking about when discussing it. Try this:
If we have a population of DNA sequences, we can imagine a case with four alleles of equal frequency. At a particular position in the DNA, one allele has A, one has C, one has G, and one has T. There is complete uncertainty about the sequence at this position. Now suppose that C has 10% higher fitness than A, G, or T (which have equal fitnesses). The usual equations of population genetics will predict the rise of the frequency of the C allele. After 84 generations, 99.9001% of the copies of the gene will have the C allele. This is an increase of information: the fourfold uncertainty about the allele has been replaced by near-certainty. It is also specified information — the population has more and more individuals of high fitness, so that the distribution of alleles in the population moves further and further into the upper tail of the original distribution of fitnesses. It's an increase in information because the uncertainty about an allele has been reduced. He isn't talking about the same thing as Dembski. You guys are not discussing the same thing when you are talking about information.
ET
April 9, 2018
April
04
Apr
9
09
2018
11:49 AM
11
11
49
AM
PDT
Glen D chimes in with:
ID doesn’t actually have anything to do with design.
And evolutionism actually doesn't have anything to do with evolution. derpET
April 9, 2018
April
04
Apr
9
09
2018
11:43 AM
11
11
43
AM
PDT
LoL! Corneel quote-mines a post- I'll help you out, corny:
Generally speaking, finding some operational way to identify who the designer is, what tools it uses, when it designs, how to distinguish design from non-design, etc.- Flint @ TSZ
That said we do have a scientific methodology for distinguishing between intelligent design and nature. And you and yours don’t have anything but “Not ID!” Your lack of integrity gets exposed when you do crap like that, corny- corny failed to mention that part of my responseET
April 9, 2018
April
04
Apr
9
09
2018
11:41 AM
11
11
41
AM
PDT
ET: Joe Felsestein, if interested, can answer my arguments. If his answer implies his concepts about CSI, he can mention them and I will try to understand if those concepts are relevant. I am not interested in his specific debate with Dembski, for the reasons I have explained.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
11:39 AM
11
11
39
AM
PDT
dazz at TSZ: I see that I had not understood well your statement quoted at #813, so my answer is obviously wrong. I apologize. Too much rush! :) The correct answer is: Fossils are interesting, but they tell us about morphology, not the molecular basis for it. So, fossils can really be useful only in the measure that we understand the molecular information related to the morphological patterns we observe.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
11:37 AM
11
11
37
AM
PDT
gpuccio- Until you read what Joe Felsenstein says about CSI you won't understand hos posts on the subject. His thinking about it is too convoluted to try to respond to unless you have that as a reference. the link is in post 782. It is an article that is posted on the NCSE's website (the alleged National Center for Science and Education)ET
April 9, 2018
April
04
Apr
9
09
2018
10:59 AM
10
10
59
AM
PDT
dazz at TSZ:
Oh, and of course, gpuccio, the saltationist result also implies that transitional fossils in vertebrate evolution would falsify your theory, don’t you think?
Of course not. that's the difference with neo-darwinism. Design does not require the expansion and fixation of each simple step. Which is instead required by neo-darwinism. Are you a little confused, or what?gpuccio
April 9, 2018
April
04
Apr
9
09
2018
10:27 AM
10
10
27
AM
PDT
dazz at TSZ:
Unfortunately that looks nothing like the kind of macro-design events that your theory requires All I see there is micro evolution
Not at all. The final transformation to an ORF is only that: the final step. But the protein sequence is prepared before that, by transposon activity. And, when it is released as a protein, it seems to be already functional. Without any intervention of NS.
I suspect we will only see gradual development if we look at what goes on inside that cocoon, should we check?
It usually happens in about a month. Not so gradual. Would you agree then that if the first vertebrate precursor of fish had emerged from a lancelet in some protected niche in the sea, by design, in a time window of about one month, you would have no problems with that?
The many ones with homologies couldn’t have possibly evolved while still functional, right?
Not by RV + NS, of course. By design, definitely yes. Not gradually. Each protein would be designed in a non functional state, and then released. With all that is necessary for its specific function.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
10:25 AM
10
10
25
AM
PDT
dazz at TSZ: April 9, 2018 at 2:09 pm I still don't agree with your views about this point (phenotypic slatation), but I will leave it to that. I don't like to repeat the same arguments. You have expressed your arguments, and I have expressed mine. But you are of course completely wrong in your final conclusions:
So what you actually have is special creation + common descent. How ironic, after all this time putting up the that old creationist tripe “evolution fails cuz cats don’t give birth to dogs” now we learn that’s pretty much how ID works.
Special creation??? Who ever spoke of creation? Of course, it is special design. With or without saltations, it's design all the same. Look, I am not a creationist at all. Not because I don't believe in creation (of course I believe in it, as my personal religious conviction). But I have never made any "creation science". Nothing in my scientific arguments is baesd on any religious idea. All that I say can be shared by anyone, whatever his ideas about religion. That sai, do as you like, and call me as you like. We live in a free will world.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
10:14 AM
10
10
14
AM
PDT
Joe Felsenstein at TSZ: April 9, 2018 at 12:48 pm I don't know what you mean by this post. Of course I accept 500 bits as a good threshold in all general cases: it certainly puts the observed result beyond any possible reach by RV alone. That is why I use it. Of course, some lower threshold would be more than enough for a biological system, which has much lower probabilistic resources than the whole universe. As I have stated very clearly in my previous answers to you (and to many others) my arguments against the powers of NS are not probabilistic. They are empirical, even if they include the probabilistic limitations of RV. Very briefly: a) Empirical data clearly show that the random emergence of a new naturally selectable function has severe limitations of complexity. Even if it is not two AAs, it is not much more. See also the many times linked paper: Waiting for Two Mutations: With Applications to Regulatory Sequence Evolution and the Limits of Darwinian Evolution http://www.genetics.org/content/180/3/1501 b) The optimization of the randomly generated function has severe limits too. In all known cases, it's a few AAs at most. c) Complex functions, like the alpha and beta chains of ATP synthase, cannot certainly emerge as simple mutations of 2 or 4 AAs by RV. And then be optimized by hundreds of single AA steps, each of them naturally selected. That has never been observed, of course, and is well beyond reason, given all that we know about available biological data. d) No naturally selectable pathway to complex functions has ever been observed, found in the lab, or even imagined in some detail. In that sense, from a scientific point of view, the existence of such pathways is a myth and nothing else. And yet, according to neo-darwinism, those pathways should be the absolute rule. This is only a brief summary. You can find the details of my arguments in my OP (and following discussion): What are the limits of Natural Selection? An interesting open discussion with Gordon Davisson https://uncommondescent.com/intelligent-design/what-are-the-limits-of-natural-selection-an-interesting-open-discussion-with-gordon-davisson/ Maybe you could have a look at them, or comment on them before stating that "gpuccio has presented no argument as to why that 500-bit limit cannot be exceeded simply by natural selection".gpuccio
April 9, 2018
April
04
Apr
9
09
2018
10:02 AM
10
10
02
AM
PDT
Corneel at TSZ:
What I am saying is that, until you show me how a protein behaves that does NOT have such jumps, I do not trust your plots to show me which proteins DO have them.
That's easy. Many of them have no jumps. For example, the beta chain of ATP synthase (an old friend) has no great jumps throughouts its metazoan history. It starts at more than 1.5 baas and gets gardually to abou 1.8 in Afrotheria. Very smoothly. Ehm, a very small jump can be seen, at the vertebrate transition. But very small. It's not my fault, after all. :) The reason why this sequence has no great jumps is rather simple: it already shows 1.25 baa in E. coli. If it ever had any big jump, that was a very, very long time ago!gpuccio
April 9, 2018
April
04
Apr
9
09
2018
09:47 AM
9
09
47
AM
PDT
Corneel at TSZ: April 9, 2018 at 11:29 am
Entropy’s example of lactate dehydrogenase demonstrating enzyme promiscuity is here. You dismissed it, but it is quite relevant to substrate specificity of E3 ubiquitin-protein ligases as well.
Not so. That case is about an enzyme who already has a specific folding potentially functional for some class of simple biochemical molecules, and for some class of reactions. A simple substitution at the active site can change (sometimes even a lot) the affinity for specific substrates in the range of possible substrates for that folding and general function. That can be seen in many cases inside a protein family, where the structure and folding are mostly shared, and so is most of the sequence specificity. Not so in the case of the specificity of E3 ligases. That specificity is about recognizing completely different target proteins, and their appropriate state. My data about homology show clearly that a great part of the protein sequence specificity is involved in that, usually a very long sequence, much longer than the domain part involved in the common ubiquitin transferase process. You are comparing two completely different scenarios.
Haha, that’s rich. So you have yourself shown how the human form of PRICKLE1 has evolved by an already functional protein acquiring an additional function? And this is not a rung of the ladder why exactly?
Not at all. Again you don't understand. The scenario is similar to that of E3 ligases. The function of the protein is modular, it is due to the interaction of (at least) two different parts of the molecule. In the case of Prikle1, there is a domain part which is more conserved in prevertebrates. IOWs the domain is older, it appeared previously. The non domain part, instead, is taxonomically restricted. In vertebrates, it is completely different from the same part in hymenoptera, for example, but it is highly conserved in each of the two groups of organisms, as I have shown here: Information jumps again: some more facts, and thoughts, about Prickle 1 and taxonomically restricted genes. https://uncommondescent.com/intelligent-design/information-jumps-again-some-more-facts-and-thoughts-about-prickle-1-and-taxonomically-restricted-genes/ As I have already said, this is an analysis of the different functional specificities of two different functional modules in the same protein. With different evolutionary histories. It has nothing to do with ladders. A ladder is a realistic pathway through which a new original complex functionv that did no exist before, appears by simple AA modifications starting from some unrelated sequence, that existed before and had some different function or no function at all, and where an initial mutation explained by RV only already provides the new function in a naturally selectable form, and all the other steps are simple and, each one of them, naturally selectable over the previous step. I hope that's clear. Neither Entropy, nor you, nor, least of all, myself, have ever provided such a ladder. Because, very simply, such a ladder does not exist.
Protein evolution will involve a certain amount of swapping, duplicating, adding and deleting functional domains. Mutations of this type do not require a designer and are valid steps on your ladder to increased functional complexity.
Re-use of domains in multi-domain proteins is a different problem. I have never used it as a scenario of functional information simply because in that scenario the measurement of the functional information is much more difficult. Indeed, in the ideal case where the sequence of the modules is always kept the same (which is not true in most cases, because they are often adapted to each new protein), the functional information involved is only that implied by the probability of getting that particular assemblage over all possible random assemblages. I believe that a designer can probably be inferred in those cases too, but you can certainly understand that there are many more variables, and too little is known to make a quantitative reasoning about that. I prefer easier tasks, which can be done with precise and explicit reasonings, to difficult analyses, that would remain vague and uncertain anywhere. That's why I stick to sequence analysis. And all my reasonings are about the building of functional sequences. Why not? We have thousands, millions of functional sequences. In functional sequences the bits are obvious, they are there to be measured. So, your statement that: "Mutations of this type do not require a designer and are valid steps on your ladder to increased functional complexity." is completely unsupported, and irrelevant to the problem of how functional sequences emerge. However, if you could demonstrate (which you have not even tried) that some modular recombination of functional parts really is in the range of RV, that would be a simple recombination. Therefore no ladder. To show that there is a ladder to complex function, you should demonstrate that: a) Joining the existing functional modules A, B and C to get a new function is a complex transition: IOWs, that it requires more than 500 bits of information. b) That it happens in many steps, each of them simple (a few bits) c) That each step from A, B, and C to A+B+C in a new functional configuration is naturally selectable over the previous step. That would be a ladder for complex domain recombination. You want to try?gpuccio
April 9, 2018
April
04
Apr
9
09
2018
09:27 AM
9
09
27
AM
PDT
Do you agree that your results show that the Designer hardly added any complex functional information to the human lineage since the human-chimpanzee split?
Question-beggingET
April 9, 2018
April
04
Apr
9
09
2018
08:59 AM
8
08
59
AM
PDT
gpuccio Corneel
Not much at the level of protein coding genes, I agree. Probably some has been added at that level too, but not much. Certainly much less than at some other transitions.
However, large changes in splicing patterns and gene expression.bill cole
April 9, 2018
April
04
Apr
9
09
2018
08:56 AM
8
08
56
AM
PDT
Corneel at TSZ:
Apropos of nothing: Do you agree that your results show that the Designer hardly added any complex functional information to the human lineage since the human-chimpanzee split?
Not much at the level of protein coding genes, I agree. Probably some has been added at that level too, but not much. Certainly much less than at some other transitions.gpuccio
April 9, 2018
April
04
Apr
9
09
2018
08:51 AM
8
08
51
AM
PDT
Too funny- Apparently the "problems" with ID have nothing at all to do with ID!:
Generally speaking, finding some operational way to identify who the designer is, what tools it uses, when it designs, how to distinguish design from non-design, etc.- Flint @ TSZ
Umm, we don't even ask those questions until AFTER (intelligent) design has been detected and is being studied. And that means those questions are irrelevant to ID, which is only about the (intelligent) DESIGN. These people have thinking issues. Thankfully not one is any kind of investigator. That said we do have a scientific methodology for distinguishing between intelligent design and nature. And you and yours don't have anything but "Not ID!"ET
April 9, 2018
April
04
Apr
9
09
2018
06:42 AM
6
06
42
AM
PDT
1 3 4 5 6 7 32

Leave a Reply