Treatments and Science - Read Only - BaldTruthTalk.com
+ Reply to Thread
Page 1 of 2 1 2 LastLast
Results 1 to 10 of 11
  1. #1
    Member
    Join Date
    May 2014
    Posts
    75

    Default Treatments and Science - Read Only

    Summary - First Post

    The original thread was becoming too big and it was becoming difficult for users to find the more important information which prompted the need for a new thread will all the research and protocols condensed into a read-only thread with titles so people can search for what they're after.

    Please do not post in this thread.

    This thread should only be used for posting research and not for any discussion whatsoever as it will only make it harder to find things for readers. If you would like to discuss anything in this thread or have any questions then please quote a post (ideally only a part) and post in the original thread linked above. I have a feeling someone is going to try to be a smartass and post in this thread anyway, so please - for everyones sake, do not post in this thread even if you have a compulsion to do so. If you would like to contribute some research then please run it by me first in the other thread so we can vigorously analyse the literature before writing up anything official. We only want research with supporting literature from multiple sources and research that is deemed important in understanding/treating AGA. You will not find bro-science or pseudo-science in here and I intend to keep it this way.

    Treatments:

    You can find all the research on the supplements/herbs and why I've recommended these dosages/vehicles, listed in this thread. If you want to search for something specific you might have to register to use the search thread function. Alternatively you can use this advanced Google search query to find posts in this thread.

    Oleuropein

    1mg-1.5mg/ml
    Vehicle: Ethanol + PG (80/20)
    Agonist: WNT10b, IGF-1, β-catenin
    Inhibits: DKK-1

    Amazon UK - Swanson Olive Leaf Extract Super Strength 750mg, 60 Capsules
    Amazon UK - Swanson Olive Leaf Extract Super Strength 750mg, 60 Capsules

    The extract above contains 150mg actual oleuropein so for a 50ml solution you'd use 1/3 of a capsule to achieve 1mg/ml. If you're going to use minox as a vehicle (which I do) then you'd use a little more than a third for 60ml of minox. Oleuropein has a fairly high molecular weight and will reduce the solubility of other molecules in the same solution so you might want to use this as a standalone mixture.

    EGCG

    5mg-15mg/ml
    Vehicle: Ethanol - PG (80/20)
    Agonist: β-catenin (via inhibition)
    Inhibits: Axin, GSK3β, AR (mild), 5ar (not EGCG)

    Amazon UK - Swanson Teavigo EGCG 150mg (actual) 30 caps
    Amazon US - Swanson Teavigo EGCG 150mg (actual) 30 caps

    You can use any brand of EGCG but I use teavigo since its got a higher concentration of EGCG. The other green tea brands will contain less EGCG but will contain other catechins that have the ability to inhibit 5ar. EGCG tends to degrade over time and degrades faster with higher humidity and temperatures so it's better to make smaller batches. You can use as much EGCG as you like but be sure not to oversaturate the solution because you'll only waste the excess. As for EGCGs AR inhibitory potency I'd guesstimate its around 30%. You can make up the rest by stacking it with a 5ar inhibitor like GLA which should be enough for the growth agonists to work properly.

    Gamma Linoleic Acid (GLA) + Linoleic Acid

    Soluble in Ethanol, Oil
    Agonist: VEGF (safflower, LA), IGF-1 (LA)
    Inhibits: 5ar, TGF-β1 (safflower)

    Sonova 400 - GLA 400mg 60 caps
    Amazon UK Solgar Borage Oil - 300mg GLA, 470mg LA 60 caps
    Amazon US Solgar Borage Oil - 300mg GLA, 470mg LA 60 caps

    I've been using Evening primrose oil mixed with minox for two weeks now and I've noticed slightly more vellus to terminal transitions. GLA/LA has alot of potential to inhibit 5ar quite potently. Borage seed extract contains more GLA than Evening primrose oil, but Sonova 400 contains almost double the amount of GLA in borage oil (with less LA: 200mg). Its quite pricy at 35$ so Borage oil is probably the better alternative. If you cant get borage oil then Evening primrose oil is an option. I would stick to capsules instead of oils as their made for oral consumption so theres very little chance they'll have additives or extra ingredients.

    Rosemary (Rosmarinus Officinalis)

    Solubility/Dose unknown
    Vehicle: Ethanol - PG (80/20)
    Inhibits: AR, 5ar

    Amazon UK - Swanson Rosemary 400mg 90 caps
    Amazon US - Swanson Rosemary 400mg 90 caps

    I havent tried Rosemary yet but it has been shown to work topically at reducing AR/5ar in mouse models that have testosterone induced alopecia. Be sure to check the other thread for any updates on my personal experiments with Rosemary.

    Apple Polyphenols (Procyanidin B2)

    Dose unknown
    Soluble in water/ethanol
    Inhibits: PKC


    Amazon UK - Swanson Apple polyphenol 125mg 60 caps

    Amazon US - Swanson Apple polyphenol 125mg 60 caps

    I am yet to use Apple polyphenols but there is strong evidence to suggest it can promote hair growth when applied topically

    Valproic Acid/Sodium Valproate

    50mg/ml (water), 30mg/ml (ethanol)
    Vehicle: Water/Distilled Water
    Agonist: β-catenin (via inhibition)
    Inhibits: GSK3β

    UnitedPharmacies US - Sodium Valproate 200mg 10 tablets
    4nrx UK - Sodium Valproate 200mg 10 tablets

    I've not used Valproic acid yet but I've ordered some and will begin using it at 25mg/ml to start off with. Its a powerful GSK3b inhibitor and will be very synergistic with minox at increasing β-catenin levels. There are reports it can be quite irritating so thats something to keep in mind.

    Polygonum Multiflorum (Fo-Ti)

    Dose unknown
    Solubility: Ethanol?
    Agonist: SHH, β-catenin, FGF-7, IGF-1

    Amazon UK Swanson Fo-Ti 500mg 60 caps
    Amazon US Swanson Fo-Ti 500mg 60 caps

    I havent used this nor have I seen any reports of it being used but I plan to test in sometime in the near future to see if it works.

    Baicalin

    Dose Unknown
    Solubility: water (<1mg/ml), ethanol (<1mg/ml) (solubility increases with temperature)
    Agonist: WNT/β-catenin

    Puerarin (Kudzu extract)

    Dose unknown
    Solubility: Ethanol (increased with temperature and physical stirring)
    Agonist: VEGF, β-catenin
    Inhibits: 5ar (flower only), AR (flower only)

    Amazon UK - Planetary Herbals Full Spectrum Kudzu Tablets, 60 caps
    Amazon US - Planetary Herbals Full Spectrum Kudzu Tablets, 60 caps

    Unfortuneately I couldnt find any extracts that contain only the flower which would have been ideal but the product I linked above has 100mg of the kudzu flower blended with the Root which isnt too bad. I havent tried this yet but the studies say its a potent AR/5ar antagonist and activates the PKA/AKT pathway which we know is good for proliferation and GSK3b inhibition.

    ----


    Vehicle

    80/20 Ethanol/Propylene Glycol

    - 190 proof grain alcohol
    not methanol or denatured alcohol/isopropyl/isopropanol

    UK: Spirytus 95% £31 + £5 Sipping - They also ship to US but its $50 for shipping
    US: Everclear if you can get it. (can only be bought in a select few states)

    - 150 proof without additional ingredients/flavourings
    If you cant find 190 proof then 75% is good enough

    Propylene Glycol

    UK: Amazon UK - Pick whichever is cheapest and is fairly high concentration
    US: Amazon US

    The science behind using Ethanol and PG is that the ethanol breaks down the skins lipid barrier and weakens the stratum corneum layers barrier function temporarily thus allowing molecules to pass through better. The propylene glycol helps keep the molecules from crystallizing and drying up and keeps soluble long enough to be absorbed. You can find the science in this thread in the Vehicles post. Try to avoid mixing high molecular weight molecules in the same solution as you'll reduce saturation of both. Using oils or creams immediately before or after the Ethanol solution might reduce ethanols lipid cutting ability - I tend to wait an hour or two before using oils.

    Please do not post in this thread. This thread is for read-only purposes. The original thread is where you can quote posts from this thread if you have any questions or would like to discuss the science.

  2. #2
    Member
    Join Date
    May 2014
    Posts
    75

    Default Androgens

    Androgens increase GSK3β:

    Androgen treatment revealed a significant decrease in the cytoplasmic/total β-catenin protein ratio and upregulation of the activity of glycogen synthase kinase-3β in DPC, indicative of canonical Wnt pathway inhibition.

    http://www.ncbi.nlm.nih.gov/pubmed/22283397
    I suspect this is due to AR's negative feedback loop found in the prostate cells, wherein GSK3β represses AR activity (controversial atm). By reducing AR this additional increase in GSK3b will be negated. This further establishes the notion that AR can reduce β-Catenin at multiple levels.

    The Androgen Receptor Antagonizes Wnt/β-Catenin Signaling in Epidermal Stem Cells

    Conversely, Filamin A (FlnA), which is required for AR activation in response to androgen stimulation (Castoria et al., 2011; Mooso et al., 2012), was significantly downregulated by β-catenin activation, an effect that was antagonized by testosterone (Supplementary Figure S5c online). The observation that testosterone increased FlnA expression in the presence of 4-OHT is consistent with the conclusion that AR signaling antagonized β-catenin signaling. As 4-OHT treatment led to a major reduction in FlnA, it is not surprising that there was no further effect of bicalutamide.

    Sustained β-catenin activation in combination with AR inhibition led to an increase in platelet-derived growth factor receptor-α-positive (Figure 5a) and Vimentin-positive (Figure 5d) fibroblasts adjacent to ectopic HFs.
    Quote Originally Posted by InBeforeTheCure View Post
    According to this, AR -> PTEN normally (?), which inhibits the Pi3K -> AKT step. But supposedly PTEN is widely expressed throughout the body as a tumor suppressor. I'm not sure how that would affect the efficacy of increasing Pi3K if AR's inhibitory effect on the Akt pathway is one step downstream of Pi3K. Any ideas?
    So this means in the presence of AR, anything that wants to activate AKT via PI3K will be unsuccessful. This includes IGF-1, shh, EGF (wiki). Beautiful diagram here: https://en.wikipedia.org/wiki/File:M...thway-v1.7.svg

    Apparently PTEN deficiency results in accelerated hair follicle morphogenesis and enhanced AKT(PKB) activation

    (Keratinocyte-specific Pten Deficiency Results in Epidermal Hyperplasia, Accelerated Hair Follicle Morphogenesis and Tumor Formation).

    Activation of the transforming growth factor α/EGFR pathway, IGF-1, or v-Ha-ras in the epidermis of transgenic mice leads to skin hyperplasia, hyperkeratosis, and tumor formation. Transgenic animals overexpressing IGF-1 also show accelerated hair growth and mice lacking IGF-1R exhibit hypoplastic skin. Because each of EGFR, IGF-R, and Ras triggers PI3′K signaling and PTEN regulates this pathway, we felt it likely that the PTEN gene would play an important role in skin development and oncogenesis.

    These condensations supply permissive and instructive signals that govern the position and type of hairs and other appendages developed. The expression of patterning genes such as those in the wnt/β-catenin/Lef-1 signaling pathway are thought to regulate these signals. Perhaps significantly, Pten has been shown to negatively regulate the β-catenin/Lef-1 pathway by inhibiting the nuclear accumulation of β-catenin and activation of Lef-1 in a prostatic cell line. However, in our hands, no definite difference in the subcellular distribution of β-catenin in k5Ptenflox/flox cells was observed (data not shown). Both k5Ptenflox/flox mice and k5IGF-1 Tg mice show accelerated hair growth at day 5, indicating that common molecules downstream of PKB/Akt in addition to β-catenin, or molecules downstream of ERK, may account for the accelerated skin morphogenesis in these mice.
    Very interesting.

    I found a study talking about synergistic effects of 5ar inibitors and AR antagonists:

    Synergistic antiandrogenic effects of topical combinations of 5 alpha-reductase and androgen receptor inhibitors in the hamster sebaceous glands

    Progesterone (P), a 5 alpha RI, and spironolactone (SL), an ARI, produced a dose responsive decrease in SGS at topical concentrations of 0.01% to 5.0%. At concentrations of 1, 3, and 5%, P and SL combinations produced neither an additive nor synergistic inhibition of SGS. At very low concentrations of up to 0.10%, neither P nor SL alone produced any effect on SGS. When combinations of these two steroids were applied at low concentrations, SGS decreased unilaterally to approximately 50%. This synergy occurred best at a P:SL ratio of 1:2. The lower effective concentrations of P may be explained by its greater percutaneous absorption. Synergy was also demonstrated at low concentrations with other antiandrogens: cyproterone acetate, canrenone, hydroxyflutamide, and N-N-diethyl-4-methyl-3-oxo-4-aza-5 alpha-androstane- 17 beta-carboxamide.
    So using both an AR suppressor and a 5ar antagonist, theoretically there should be even greater AR suppression. EGCG can inhibit AR to an extent, but when DHT binds to AR its effects are order of magnitudes greater than T. I found out that Gamma Linoleic acid can inhibit 5ar quite well, you can read more about GLA in a separate post I made in this thread.

  3. #3
    Member
    Join Date
    May 2014
    Posts
    75

    Default DKK-1 - Dickkopf-1

    DKK-1 has been known for a while to be an important player in the development of AGA but it has been largely ignored by many people with most of our attention diverted towards newer findings like PGD2 and PGE2.

    In short DKK-1 antagonizes the LRP5/6 receptors which the WNTs require in order to exert their functions.

    https://en.wikipedia.org/wiki/DKK1

    The dickkopf protein encoded by DKK1 is an antagonistic inhibitor of the WNT signaling pathway that acts by isolating the LRP6 co-receptor so that it cannot aid in activating the WNT signaling pathway.[4] DKK1 was also demonstrated to antagonize the Wnt/β-catenin pathway via a reduction in β-catenin and an increase in OCT4 expression.[5]
    Not only that but DKK1 is involved in the β-catenin/TCF negative feedback loop which prevents aberrant cellular proliferation.

    DKK1, a negative regulator of Wnt signaling, is a target of the beta-catenin/TCF pathway.

    Wnt signaling plays an important role in embryonic development and tumorigenesis. These biological effects are exerted by activation of the beta-catenin/TCF transcription complex and consequent regulation of a set of downstream genes. TCF-binding elements have been found in the promoter regions of many TCF target genes and characterized by a highly conserved consensus sequence. Utilizing this consensus sequence, we performed an in silico screening for new TCF target genes. Through computational screening and subsequent experimental analysis, we identified a novel TCF target gene, DKK1, which has been shown to be a potent inhibitor of Wnt signaling. Our finding suggests the existence of a novel feedback loop in Wnt signaling.
    Here's a study showing how DHT increases DKK1 via P53 and ROS generation.

    Dihydrotestosterone-inducible dickkopf 1 from balding dermal papilla cells causes apoptosis in follicular keratinocytes.

    Recent studies suggest that androgen-driven alteration to the autocrine and paracrine factors produced by scalp dermal papilla (DP) cells may be a key to androgen-potentiated balding. Here, we screened dihydrotestosterone (DHT)-regulated genes in balding DP cells and found that dickkopf 1 (DKK-1) is one of the most upregulated genes. DKK-1 messenger RNA is upregulated in 3-6 hours after 50-100 nM DHT treatment and ELISA showed that DKK-1 is secreted from DP cells in response to DHT. A co-culture system using outer root sheath (ORS) keratinocytes and DP cells showed that DHT inhibits the growth of ORS cells, and neutralizing antibody against DKK-1 significantly reversed the growth inhibition of ORS cells. Analysis of co-cultured ORS cells showed a significant increment of sub-G1 apoptotic cells in response to DHT. Also, recombinant human DKK-1 inhibited the growth of ORS cells and triggered apoptotic cell death. In addition, DHT-induced epithelial cell death in cultured hair follicles was reversed by neutralizing DKK-1 antibody. Moreover, immunoblotting showed that the DKK-1 level is up in the bald scalp compared with the haired scalp of patients with androgenetic alopecia. Altogether, our data strongly suggest that DHT-inducible DKK-1 is involved in DHT-driven balding.
    Other possible leads I came across include Vitamin D3 and its target receptor VDR::

    The Wnt antagonist DICKKOPF-1 gene is induced by 1a,25-dihydroxyvitamin D3
    associated to the differentiation of human colon cancer cells


    The Wnt–b-catenin pathway is aberrantly activated in most colon cancers. DICKKOPF-1 (DKK-1) gene encodes an extracellular Wnt inhibitor that blocks the formation of signalling receptor complexes at the plasma membrane. We report that 1a,25- dihydroxyvitamin D3 [1,25(OH)2D3], the most active vitamin D metabolite, increases the level of DKK-1 RNA and protein in human SW480-ADH colon cancer cells. This effect is dose dependent, slow and depends on the presence of a transcription-competent nuclear vitamin D receptor (VDR). Accordingly, 1,25(OH)2D3 activates a 2300 bp fragment of the human DKK-1 gene promoter. Chromatin immunoprecipitation assays revealed that 1,25(OH)2D3 treatment induced a pattern of histone modifications which is compatible with transcriptionally active chromatin. Exogenous expression of E-cadherin into SW480-ADH cells results in a strong adhesive phenotype and a 17-fold increase in DKK-1 RNA. In contrast, an E-cadherin blocking antibody inhibits 1,25(OH)2D3-induced differentiation of SW480-ADH cells and DKK-1 gene expression. Remarkably, in vivo treatment with the vitamin D analogue EB1089 induced DKK-1 protein expression in SW480-ADH cells xenografted in immunodeficient mice, and a correlation was observed in the expression of VDR and DKK-1 RNA in a series of 32 human colorectal tumours. These data indicate that 1,25(OH)2D3 activates the transcription of the DKK-1 gene, probably in an indirect way that is associated to the promotion of a differentiated phenotype. DKK-1 gene induction constitutes a novel mechanism of inhibition of Wnt signalling and antitumour action by 1,25(OH)2D3.
    It appears VDR (Vitamin D receptor) mediates DKK-1 protein expression in the presence of E cadherin by binding to a promoter region on Dkk1 gene.
    Furthermore, DHT has been shown to upregulate VDR via ERBeta!

    Sex steroids induced up-regulation of 1,25-(OH)2 vitamin D3 receptors in T 47D breast cancer cells.

    Abstract
    There is evidence indicating that 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] through binding to its specific receptor (VDR) exerts an antiproliferative effect on breast cancer cells. Considering the importance of receptor regulation in modulating the target cell responsiveness to hormones, the effect of dihydrotestosterone (DHT) and estradiol-17 beta (E2) on the regulation of VDR number was investigated in T 47D human breast cancer cells that also express androgen and estrogen (ER) receptors. Exposure to 10(-7) M DHT for 72 h resulted in a significant increase in VDR levels. Similar results were obtained with 10(-7) M E2. DHT- and E2-induced up-regulation was completely suppressed by 10(-6) M tamoxifen (TAM) addition but unaffected by 10(-6) M flutamide. TAM treatment alone produced a significant dose-dependent increase in VDR content, that was maximal at 10(-6) M. Our data strongly suggest, for the first time, an up-regulation of VDR by DHT and E2 via an ER-mediated mechanism.
    Although this study’s focus is on breast cancer cells, it’s still points us in the right direction.[/QUOTE]

    Vitamin D has been shown to have a biphasic effect on hair growth:

    http://www.ncbi.nlm.nih.gov/pubmed/8077696

    At higher concentrations of 1,25(OH)2D3, there was a dose-dependent inhibition of both follicle and fiber growth (IC50 values of 100 nM), in part due to reduction in the growth periods. There was a marked delay between the onset of 1,25(OH)2D-induced hair follicle and hair fiber growth inhibition.
    This exactly inline with the oxford study:

    Time-course and dose-curve experiments showed that 1,25(OH)2D3 (107 M) caused a slow 3- to 5-fold induction of DKK-1 RNA at 24–48 h upon treatment. The effect of 1,25(OH)2D3 was specific, as several hormones (dexamethasone, retinoic acid, progesterone and oestradiol) acting through members of the superfamily of nuclear receptors similar to VDR did not induce DKK-1. The induction of DKK-1 was confirmed at the protein level and in another colon cancer cell line. Immunofluorescence studies confirmed the increase in DKK-1 protein expression following 1,25(OH)2D3 exposure and showed its preferential localization in the cell periphery, Golgi apparatus and vesicles of the exocytic route. These results confirmed that 1,25(OH)2D3 induces DKK-1 expression with slow kinetics, which precluded the use of translation inhibitors such as cycloheximide to investigate whether the induction is direct or indirect.
    Here's a diagram I made to illustrate how these pathways contribute to AGA (the estrogen receptor Beta pathway is incorrect given recent findings which you can read more about in this thread).



    I've been doing some more research and I came across some new information on DKK-1. We know that oxidative stress (ROS) induces the expression of DKK1, but until now I didnt know how that happened. This excerpt sheds some light on this matter:

    The oxidative stress response regulates DKK1 expression through the JNK signaling cascade in multiple myeloma plasma cells

    To investigate the upstream stimuli responsible for JNK pathway activation and DKK1 up-regulation by lenalidomide, we analyzed changes in gene expression that correlated with DKK1 up-regulation following lenalidomide treatment in vivo. Microarray analysis showed a clear indication of the activation of genes involved in the cellular response to oxidative stress, which is in line with previous reports indicating that thalidomide could induce oxidative injury in the rabbit model system by forming free radical-initiated reactive oxygen species. Interestingly, in the 40 most highly correlated genes, we identified genes encoding caspase 4 (CASP4, 8.7-fold increase; most up-regulated gene) and calpain 2 (CAPN2, 5.3-fold increase), which are involved in endoplasmic reticulum stress-induced apoptosis, and glutaredoxin (GLRX, 7.45-fold increase), glutathione peroxidase (GPX1, 5.7-fold increase), and glutathione S-transferase (GSTO1, 5-fold increase), which play important roles in cellular detoxification, suppressing activities of JNK and its upstream kinases through direct in,eractions.

    Therefore, to determine whether oxidative stress plays a role in mediating lenalidomide-induced DKK1 up-regulation, we administered the drug along with the antioxidant agent PBN. As shown in Figure 3A, pretreating cells with PBN significantly reduced lenalidomide-induced DKK1 up-regulation (expression reduced 1.5- to 3.3-fold; mean: 2 ± 0.1; P < .01). Interestingly, preincubating cells with PBN alone could reduce the basal level DKK1 expression (data not shown). Furthermore, in line with previous reports, PBN counteracted c-Jun–binding activation (P < .01; Figure 3B) and JNK phosphorylation (data not shown). Taken together, these data demonstrate that oxidative stress is an upstream stimulus responsible for JNK pathway activation and DKK1 up-regulation induced by lenalidomide.
    The antioxidant PBN could prevent the oxidative stress induced DKK-1. Just like Ascorbic Acid. Oleuropein is also a free radical scavenger. But the interesting point here is that, antioxidants could reduce DKK-1 below basal levels without a stress response stimulus. This explains why oleuropein reduced DKK-1 in the mice study. In the presence of testosterone, anti-oxidants will probably only keep DKK-1 at basal levels, but if AR is suppressed significantly, DKK-1 can be further reduced thus allowing WNT to freely bind whenever and however it wants! It seems DKK-1 is a solved case, its now time to find agents that can activate the canonical WNT pathway.

  4. #4
    Member
    Join Date
    May 2014
    Posts
    75

    Default Estrogens - ERα - ERβ - 3β Diol

    Estrogen acts through two receptors, ERα and ERβ. These receptors have different roles - sometimes even contradictory, depending on the tissue localisation and agonist binding affinity. Estrogen is not the only hormone that bind to these receptors, there can be other agents with much higher binding affinities and much much powerful activation. 3β Diol is one of those agents widely present in male prostatic tissues. It has the ability to activate ERβ very potently even moreso than Estrogen itself. 3β Diol is actually a DHT derivative, in males, since we have less Estrogen floating around, the body has developed alternative ways of activating the estrogen receptor signalling pathway to circumvent the pro-carcinogenic effects of androgens of the prostate and it would seem the scalp too.

    In addition, 3β-adiol is considered a powerful DHT metabolite since its intraprostatic protein level is 100-fold higher than that of estradiol (E2) (29). Notably, 3β-adiol has antiproliferative actions which are not reproduced by 17β-estradiol (30).

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4212017/
    We found that 3beta-Adiol not only inhibits PC3-Luc cell migratory properties, but also induces a broader anti-tumor phenotype by decreasing the proliferation rate, increasing cell adhesion, and reducing invasive capabilities in vitro. All these 3beta-Adiol activities are mediated by ERbeta and cannot be reproduced by the physiological estrogen, 17beta-estradiol, suggesting the existence of different pathways activated by the two ERbeta ligands in PC3-Luc cells.

    http://www.ncbi.nlm.nih.gov/pubmed/20562232
    My initial conclusion was that ERβ was a bad pathway but I was incorrect. Turns out ERβ has differential effects when activated by different agents, and in the case of 17β Estradiol it actually promoted fronto-temporal hair growth in males. Read below

    One clear frustration when analyzing the biochemichal pathways of Estrogen is the massive discrepancy of ER mediated pathways between genders and species. In females, ERβ in the hair follicles has a completely different distribution to males, and actually inhibits hair growth. The same in male mice. ERα inhibits hair growth and ERβ keeps ERα in check.

    The wide distribution of ERβ in human pilosebaceous unit suggests that estrogens play an important role in the maintenance and the regulation of the hair follicle and provides further evidence for estrogen action in nonclassic target tissues. Recently, it was reported that in cultured dermal papilla cells from nonbalding male donors, both ERα and ERβ showed a consistently higher expression, both at the RNA and protein levels, in occiput dermal papilla cells compared with vertex dermal papilla cells. With respect to ERβ immunoreactivity, we found that, in anagen VI follicles microdissected from frontotemporal skin, there was a remarkable distribution difference between male and female hair follicles from frontotemporal scalp skin: ERβ immunoreactivity was found in male scalp hair follicles predominantly in the matrix keratinocytes, whereas in female hair follicles, ERβ immunoreactivity was predominantly found in the dermal papilla fibroblasts. These data not only highlight substantial, previously underappreciated sex-dependent differences in ERβ expression of an important peripheral E2 target organ, but also underscore the importance of investigating whether E2 effects on the human hair follicle are location-dependent, as is well-recognized for the paradoxical hair growth effects of androgens.

    Conflicting data have been presented concerning ER expression patterns in murine hair follicles. It has been reported that ERα was expressed only in the dermal papilla and outer root sheath of telogen and early anagen mouse hair follicles and that ERβ was undetectable. Recently, however, we could show that both ERα and ERβ as well as the splice variant ERβ ins are expressed throughout the entire, depilation-induced murine hair cycle at both the protein and RNA levels. In addition, hair follicles in late anagen (anagen VI) were highly sensitive to regulation by topically applied E2, which rapidly induced premature catagen entry. Therefore, anagen VI mouse pelage hair follicles must express fully functional ERs.

    ERα immunoreactivity peaks in murine telogen follicles within the dermal papilla and the sebaceous gland, whereas the inner root sheath and outer root sheath show weaker immunoreactivity. In anagen VI, ERα immunoreactivity (IR) is detectable in the outer root sheath and the dermal papilla, whereas in early catagen it is restricted to the dermal papilla and the secondary hair germ. In anagen VI follicles, ERβ is weakly positive in hair matrix and outer root sheath, whereas in catagen and telogen follicles, ERβ is expressed in the dermal papilla, inner root sheath, outer root sheath, and the sebaceous gland. By RT-PCR, ERα and ERβ transcripts can be detected in telogen, anagen V and VI, and late catagen skin mRNA extracts. Investigation of ERβ knockout mice showed an accelerated catagen development along with an increase in the number of apoptotic hair follicle keratinocytes. Taken together, this suggests that the catagen-promoting properties of E2 in murine skin are mediated by ERα and that ERβ mainly functions as a silencer of ERα action in murine hair biology. (An additional list on reported expression of estrogen signaling components is provided in Table 2).

    http://press.endocrine.org/doi/full/...0/er.2006-0020
    This study found further discrepancies in In-Vitro and In-Vivo effects of ER Beta:

    Estrogens and Human Scalp Hair Growth—Still More Questions than Answers

    We found only two corresponding reports in the published literature, one using human anagen hair follicles from an unspecified scalp skin location of what appears to be two male individuals aged 17 and 35 y (Kondo et al, 1990), and one recent meeting abstract based on the use of female occipital scalp skin follicles (Nelson et al, 2003). Both studies report that E2 (Kondo et al: 18 nM; Nelson et al: 10 nM), significantly inhibits human scalp hair shaft elongation in vitro.
    Here theyre saying E2 inhibted hair in-vitro. This only adds to the confusion. They also did a study themselves on female hair derived from occipital regions:

    Recently, we have also studied the effects of E2 (1 nM–1 muM, Sigma St. Louis, MO) on female occipital scalp hair follicles, and have essentially confirmed hair shaft elongation-inhibitory properties of E2, which were maximal at 1 muM
    Paradoxically, Estrogens can actually increase hair growth in male fronto temporal regions (temples):

    Effect of estrogens on skin aging and the potential role of SERMs

    Recent in vitro studies have shown that 17β-estradiol inhibits female scalp hair shaft elongation (Nelson 2006), although stimulation occurs in hair follicles derived from frontotemporal male scalp (Conrad et al 2004). In addition, in female hair follicles the phytoestrogen, genistein inhibits hair shaft elongation to a similar extent as 17β-estradiol. Since genistein preferentially binds to ERβ, this opens the possibility that the inhibition of hair growth in response to 17β-estradiol may be mediated via ERβ rather than ERα (Nelson 2006). Therefore the development of selective estrogen receptor ligands may provide important clinical applications for the prevention and treatment of disorders of hair growth.
    So in males, ERβ increases frontotemporal hair growth?

    ERβ was the major steroid receptor expressed in human skin. It was highly expressed in epidermis, blood vessels and dermal fibroblasts, in contrast to ERα and AR. In the hair follicle, ERβ expression was localized to nuclei of outer root sheath, epithelial matrix and dermal papilla cells, in contrast to ERα, and the AR, which was only expressed in dermal papilla cells.
    I've not seen a single study talk about the effect of Estrogen on hair derived from female fronto temporal regions but I suspect it actually increases hair growth as evident by the typical rounded hairlines of most women.

    Studies say that estrogen prolongs anagen but inhibits hair shaft elongation (in females). Perhaps ERβ in the ORS is inhibiting proliferation, but ERα in the DPC is maintaining β-Catenin. But Estrogen acts differently in frontotemporal regions? Females have a differential scalp response phenotype. Males start off as females in the womb, so what if Androgens exploit this localised response in a detrimental manner? This sets off the chain reaction whereby follicles release DKK-1 and TGF-β1 into the ECM affecting nearby follicles that arent fully susceptible to AR's effects. Anyways, I want to know what role ERα has in male DPC. ERβ can increase proliferation of existing hair, but does it secrete growth factors in a paracrine manner? Or does ERα promote WNT activation by itself in DPC to prolong anagen?

    I think we should investigate the effects of topical Estrogen receptor agonists for hair growth seeing as the science supports the notion that Estrogens can increase temple hair growth.

  5. #5
    Member
    Join Date
    May 2014
    Posts
    75

    Default β-Catenin

    This post will be all about β-catenin, how it works to promote hair growth and how we can increase it

    I'll start off with this study that demonstrates the ability of β-catenin to induce new hair follicles:

    Transient activation of beta-catenin signalling in adult mouse epidermis is sufficient to induce new hair follicles but continuous activation is required to maintain hair follicle tumours.

    When beta-catenin signalling is disturbed from mid-gestation onwards lineage commitment is profoundly altered in postnatal mouse epidermis. We have investigated whether adult epidermis has the capacity for beta-catenin-induced lineage conversion without prior embryonic priming. We fused N-terminally truncated, stabilised beta-catenin to the ligand-binding domain of a mutant oestrogen receptor (DeltaNbeta-cateninER). DeltaNbeta-cateninER was expressed in the epidermis of transgenic mice under the control of the keratin 14 promoter and beta-catenin activity was induced in adult epidermis by topical application of 4-hydroxytamoxifen (4OHT). Within 7 days of daily 4OHT treatment resting hair follicles were recruited into the hair growth cycle and epithelial outgrowths formed from existing hair follicles and from interfollicular epidermis. The outgrowths expressed Sonic hedgehog, Patched and markers of hair follicle differentiation, indicative of de novo follicle formation. The interfollicular epidermal differentiation program was largely unaffected but after an initial wave of sebaceous gland duplication sebocyte differentiation was inhibited. A single application of 4OHT was as effective as repeated doses in inducing new follicles and growth of existing follicles. Treatment of epidermis with 4OHT for 21 days resulted in conversion of hair follicles to benign tumours resembling trichofolliculomas. The tumours were dependent on continuous activation of beta-catenin and by 28 days after removal of the drug they had largely regressed. We conclude that interfollicular epidermis and sebaceous glands retain the ability to be reprogrammed in adult life and that continuous beta-catenin signalling is required to maintain hair follicle tumours.
    The formation of hair follicles during embryogenesis depends on a series of signals that are exchanged between the epidermis and the underlying dermis. In the embryo the initiating signal comes from the dermis and involves activation of Wnt signalling; the response in the overlying epithelium also involves activation of β-catenin. In K14ΔNβ-cateninER skin the dermal signal is not required and activation of β-catenin signalling in the epidermis leads to organisation of a dermal papilla (Fig. 3G-J). In each location where new follicles formed in K14ΔNβ-cateninER epidermis there was induction of Shh, Ptc and Lef1. Just as Shh drives anagen, Shh is downstream of Wnt signalling in hair follicle development: in mice lacking Shh hair follicle formation is initiated and the dermal condensate is formed but mature hair follicles fail to develop. Lef1 is a known transcriptional target of β-catenin, required for normal hair follicle formation. Although during normal hair placode formation expression of Lef1 is regulated by Noggin, produced by dermal cells, in our system Lef1 upregulation is independent of a pre-existing dermal signal.
    So β-catenin, when elevated to significant levels can create new hair follicles by physically promoting the formation of a dermal papilla regardless of the hair follicle cycle / local signalling feedback loop control. β-catenin has been known to promote hair growth for a while now, but it was unclear as whether it could create new follicles in the absence of DPC. This new evidence suggests that β-catenin may be even more important than we thought.

    How do we increase β-catenin?

    β-catenin is normally degraded in the cell until WNT's bind to the LRP5/6 receptors, preventing the destruction complex from successfully forming. (DKK-1 inhibits LRP5/6)

    Canonical WNT/β-catenin pathway

    The canonical Wnt pathway (or Wnt/β-catenin pathway) is the Wnt pathway that causes an accumulation of β-catenin in the cytoplasm and its eventual translocation into the nucleus to act as a transcriptional coactivator of transcription factors that belong to the TCF/LEF family. Without Wnt signaling, the β-catenin would not accumulate in the cytoplasm since a destruction complex would normally degrade it. This destruction complex includes the following proteins: Axin, adenomatosis polyposis coli (APC), protein phosphatase 2A (PP2A), glycogen synthase kinase 3 (GSK3) and casein kinase 1α (CK1α). It degrades β-catenin by targeting it for ubiquitination, which subsequently sends it to the proteasome to be digested. However, as soon as Wnt binds Fz and LRP5/6, the destruction complex function becomes disrupted. This is due to Wnt causing the translocation of the negative Wnt regulator, Axin, and the destruction complex to the plasma membrane. Phosphorylation by other proteins in the destruction complex subsequently binds Axin to the cytoplasmic tail of LRP5/6. Axin becomes de-phosphorylated and its stability and levels are decreased. Dsh then becomes activated via phosphorylation and its DIX and PDZ domains inhibit the GSK3 activity of the destruction complex. This allows β-catenin to accumulate and localize to the nucleus and subsequently induce a cellular response via gene transduction alongside the TCF/LEF (T-cell factor/lymphoid enhancing factor)[16] transcription factors.
    Heres a picture to help visualise whats going on.



    Wnt Signaling through Inhibition of β-Catenin Degradation in an Intact Axin1 Complex

    The proteins surrounding β-catenin are involved in the destruction of β-catenin. First CKI prepares β-catenin for GSK3β. Once GSK3β has phosphorylated β-catenin, Axin and APC form a scaffold that that enables the β-catenin to be degraded. If any of these proteins (GSK3β, Axin, CKO) are inactivated then β-catenin will not be destroyed and will be able to translocate into the nucleus and exert its effects with the help of TCF4. WNTs like WNT10b - which oleuropein upregulates - protect β-catenin from destruction:

    The removal of β-catenin from the destruction complex is accomplished simply by direct degradation by the proteasome. This step recycles the destruction complex for another round of β-catenin degradation. We show that Wnt receptor-ligand interaction leaves the destruction complex compositionally unchanged and does not affect the activity of its kinases. The only change that we observe is the association of Axin1 with phosphorylated Lrp6 and the dissociation of β-TrCP. Indeed, phosphorylated β-catenin—still bound to the Axin1 complex—is no longer ubiquitinated and degraded. It saturates and thus effectively inactivates the Axin1 complex. We have previously reported that only β-catenin that is newly synthesized after initiation of the Wnt signal is signaling competent (Staal et al., 2002). This notion is in agreement with our current model, which predicts that newly synthesized, nonphosphorylated β-catenin will be stable in a free cytosolic form once the destruction complex is saturated. It can then translocate to the nucleus to associate with TCF and activate the Wnt transcriptional program.
    Anything that blocks Axin or GSK3β or any of the proteins required to destroy β-catenin will lead to increased β-catenin. Thats why GSK3β inibitors like Valproic acid work topically.

    It turns out activating β-Catenin pathway triggers a feedback loop increasing Axin2 (minoxidil study) (another). Axin2 can substitute axin1 and I'm not sure if the prostaglandin receptors or even EGCG can reduce Axin2 expression. Without Axin2, you'd have sustained β-Catenin level which could become cancerous in sensitive cell types. For hair this would translate to ridiculous hair regrowth rates. (More reading)

    Axin1 is the rate limiting factor for β-Catenin degradation, so if its substituted with Axin2, you're back at square one - β-Catenin will be destroyed. But if you reduce GSK3β, regardless of Axin1/2, β-Catenin will stay elevated.

    Indirubin from Angelica sinensis extract - Dong Quai can inhibit GSK3b. Another study.

    Prostaglandin E2 has been shown to increase β-catenin by interfering with the destruction complex, which is probably how it exerts hair growth promoting abilities. I realise PGE2 isnt that easy to come by and its diffiult to increase endogenous production.

    Thats where EGCG comes in: Not only does EGCG down regulate Axin expression, it also phosphorylates (inactivates) GSK3β leading to increased β-catenin levels. However I do not know how strong these effects are in comparison to PGE2.

    I've made a post on EGCG which you can find in this thread.


    PKA inactivates GSK3b expressio
    n. Agents that increase cAMP mediated PKA will inhibit GSK3b. This inlcludes IGF-1, PGE2, EGF and just about any mitogenic cytokine.

    Effects of the cAMP-elevating agents cilostamide, cilostazol and forskolin on the phosphorylation of Akt and GSK-3beta in platelets.

    PTH/cAMP/PKA signaling facilitates canonical Wnt signaling via inactivation of glycogen synthase kinase-3beta in osteoblastic Saos-2 cells.

    Western blotting demonstrated that GSK-3beta was rapidly phosphorylated (inactivated) at Ser(9) on treatment with PTH or forskolin, leading to its inactivation. Moreover, overexpression of a constitutively active mutant of GSK-3beta abolished the TCF-dependent transactivation induced by forskolin. On the other hand, overexpression of the Wnt antagonist Dickkopf-1 (DKK1) failed to cancel the effects of forskolin on the canonical Wnt pathway
    So why hasnt anyone tried forskolin yet?

    Evidence that activation of protein kinase A inhibits human hair follicle growth and hair fibre production in organ culture and DNA synthesis in human and mouse hair follicle organ culture.

    Human hair follicles were isolated from facial skin by microdissection...Human hair follicle growth was inhibited by the phosphodiesterase inhibitors 3-isobutyl-1-methylxanthine and Ro 20-1724, and by the adenylate cyclase activator, forskolin.
    Interpret that however you wish. Apparently PGE2 should inhibit facial hair growth too.

    This study shows procyanidins (found in apple skin) paired with forskolin increases hair growth rate.

    Several selective protein kinase C inhibitors including procyanidins promote hair growth.

    Forskolin, an adenylate cyclase activator, promotes hair epithelial cell growth and boosts the growth-promoting effect of procyanidin B-2. It is speculated that the hair-growing activity of procyanidins is related to their protein kinase C-inhibiting activity.
    PI3K is a known upstream mediator of AKT signalling so what about herbs that activate Pi3K? I know puerarin activates Pi3K quite strongly, so I googled, and whaddayaknow!

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3765709/
    These observations indicated that puerarin induced the phosphorylation of AKT at serine 473 and subsequently activated the phosphorylation of GSK-3β at serine 9, leading to GSK-3β inhibition and Wnt/β-catenin signaling.
    http://www.healthmonthly.co.uk/swans...FYMIwwodQfgN3Q

    Puerarin's solubility in water can be increased with higher temperatures. Adding in a starch and using steel balls to mix the solution ridiculously enhances bioavailability: study

    Puerariae Flos (the flowers of Pueraria thomsonii) was found to inihibit 5ar and grow hair in mice.

    Not sure where to get this specfic extract since kudzu is broad and we need that flower specifically to inhibit AR, but the standard swanson kudzu I linked should inhibit GSK3b - in theory.

    On the topic of inhibiting GSK3β, I'd like to briefly go over Valproic and Lithium Chloride.

    Valproic Acid Induces Hair Regeneration in Murine Model and Activates Alkaline Phosphatase Activity in Human Dermal Papilla Cells

    You can see in this figure that VA was marginally better than minox at increasing the ratio of hairs in middle anagen. Since minox increases β-catenin through a different pathway, stacking them both together would be an incredibly powerful combination.

    These results indicate that the hair-inducing activity of EBA may be independent of the Wnt/β-catenin pathway, and in fact we confirmed that EBA induced activation of Erk and Akt, which are in turn involved in keratinocyte proliferation. Interestingly, VPA induced expression of the hair follicular stem cell markers ketatin 15 and CD34 during hair formation and wound-induced growth. VPA is known to induce CD34 expression and enhance stemness. The bald scalps of men with androgenetic alopecia lack CD200-rich, CD34-positive hair follicle progenitor cells, and have a defect in conversion of hair follicle stem cells to progenitor cells, which play a role in the pathogenesis of androgenetic alopecia.
    It appears LiCl isnt too suitable for use due to its aberrant epidermis thickening effects which wasnt seen in VA. I'm on the fence with LiCl.

    However, the thickness of the epidermis was increased in skin treated with BeCl2 or PBA compared to control skin, as previously described for LiCl application. The expression of filaggrin and loricrin was abnormally increased by application of BeCl2, similar to the effect of LiCl (Figure S6B).
    For people that cant use minox then Valproic Acid seems like a better alternative with equal efficacy.

    So you can see there are quite a few ways to increase β-Catenin, be it via inhibition of GSK3β or PGE2 upregulation, you cannot grow hair without β-Catenin.

  6. #6
    Member
    Join Date
    May 2014
    Posts
    75

    Default PGE2 - Prostaglandin E2

    Its quite old knowledge that PGE2 can dramatically increase hair growth as evident by the effectiveness of bimatoprost and latanoprost at increasing eyelash growth (prostaglandin analogues). Wound induced hair growth is also driven by an increase in PGE2 so its very clear PGE2 is a very powerful agent for hair growth. So far there's been little studies outlining the exact mechanism of PGE2's hair growth abilities. Recent findings have shed light on this mystery, read on:

    Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-catenin signaling axis.

    We show that PGE2 stimulates colon cancer cell growth through its heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor, EP2, by a signaling route that involves the activation of phosphoinositide 3-kinase and the protein kinase Akt by free G protein betagamma subunits and the direct association of the G protein alphas subunit with the regulator of G protein signaling (RGS) domain of axin. This leads to the inactivation and release of glycogen synthase kinase 3 beta from its complex with axin, thereby relieving the inhibitory phosphorylation of beta-catenin and activating its signaling pathway. These findings may provide a molecular framework for the future evaluation of chemopreventive strategies for colorectal cancer.
    PGE2 inhibits Axin - which is necessary for the degradation of β-Catenin. There's not alot of drugs/treatments that can inhibit Axin, and seeing as Axin is the rate limiting factor of β-Catenin destruction, its no surprise PGE2 is a beast. Not only that but PGE2 directly activates PI3K/AKT which is known to inhibit GSK3β and as result leads to further increased β-Catenin.

    Prostaglandin E2 Stimulates the β-Catenin/T Cell Factor-dependent Transcription in Colon Cancer

    PGE2 Stimulated GSK-3 Phosphorylation—PGE2 increases the phosphorylation of GSK-3α in human embryonic kidney cells that ectopically express E-prostanoid receptors (30) and in human neuronal cells (31). The phosphorylation of GSK-3 inhibits its kinase activity, which is required for phosphorylation and degradation of β-catenin (32). To determine whether PGE2 increased the phosphorylation of GSK-3 in colon cancer cells, LS-174T cells were treated with PGE2. Levels of phosphorylated GSK-3α were rapidly elevated (Fig. 4A). An increase in the level of β-catenin was detected as well.
    PGE2 does this via the EP2 receptor and also the EP3/4 receptors:

    Prostaglandin E2 promotes human cholangiocarcinoma cell proliferation, migration and invasion through the upregulation of β-catenin expression via EP3-4 receptor.


    PGE2 is quite difficult to increase and analogues are very expensive. Castor oil is a viable alternative however. The only other agent I've found that can inhibit Axin and GSK3β is EGCG. I've made a separate post on this supplement and I personally use it topically.

  7. #7
    Member
    Join Date
    Dec 2015
    Posts
    35

    Default

    Hi chemical. Its amazing all these research that you are doing. I wanted to know if you have had significant improvement in your hair using your findings??
    thank you

  8. #8
    Member
    Join Date
    May 2014
    Posts
    75

    Default Pgd2 - ptgds

    @UNBEAT please use the other thread for questions and discussion.

    Quote Originally Posted by InBeforeTheCure View Post

    Interestingly enough, PGD2 activation of CRTH2 supposedly activates GSK3, and also results in phosphorylation of AKT.





    Link: New Drugs and Targets for Asthma and COPD
    Huge thanks to InBeforeTheCure for this contribution.

    So far I've been struggling any literature on the physiological effects of GPR44/CRTH2 on hair follicles but this finding is a major leap in our understanding of what PGD2 actually does. If you find my post on β-catenin you can see that GSK3β activation enhances the degradation of β-catenin so anything that increase GSK3β will retard hair growth significantly. This effect of GPR44, I suspect is due to its Pi3K/AKT(PKB) phosphorylating (inactivating) properties. Pi3K/PKA reduces GSK3β so by inhibiting PKA, PGD2 can enhance the degradation of β-Catenin.

    Quote Originally Posted by InBeforeTheCure View Post
    You forgot the link: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3319975/

    Not only that, but PGD2 production is 12 times higher in bald vs. haired scalp in men with AGA:

    http://www.ncbi.nlm.nih.gov/pmc/arti...975/figure/F2/
    Quote Originally Posted by Chemical View Post
    Figure 2E is the averaged level of PGD2 from a greater sample, using a more accurate measurement method. Not as high as 12x but still high enough to retard hair growth. Paired with AR it will definitely cause balding.
    Quote Originally Posted by InBeforeTheCure View Post
    Interestingly enough, not everyone with hair loss is sensitive to PGD2's effects. A poster from the World Congress for Hair Research last month, courtesy of hellouser (right click and select "view image" or "open image in new tab" for a larger version):

    poster

    As you can see, they identified a few SNPs (which appear to be in perfect linkage disequilibrium) in the CRTH2 (GPR44) gene associated with PGD2 sensitivity as well. I would guess that PGD2 is one problem (in those people who are sensitive to it), but not the only problem. Maybe PGD2 sensitivity accelerates things?
    Quote Originally Posted by Chemical View Post
    "Approx half of Alopecia patients" - random sample and half were immune? My stats knowledge isnt the best but this is a clear indication that PGD2 isnt the holy grail after all. I would also like to see more research on genetic influence. It could be that we've all got different variations of AGA and respond only when the main individual-specific antagonists are suppressed.
    Quote Originally Posted by Chemical View Post
    PTGDS is being upregulated as anagen progresses, I cant figure out how. One theory is that its TCF/LEF dependent like Axin2 is, but is it an extracellular process? If so, then human hair should enter telogen in groups. Which isnt the case because they cycle independently. Furthermore:

    Firstly, the first two cycles of the mouse hair follicle are synchronized whereas in humans at a time when biopsies could be taken, neighbouring follicles cycle independently of each other. Secondly, the mouse hair cycle is short, taking about 3 weeks; in contrast, human scalp hairs have a cycle time of several years, and even vellus hairs take months. The short synchronized hair cycle thus allows hair follicles to be harvested and examined at specific time points in the cycle very easily.

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1571051/
    The mouse model isnt entirely reliable for analysis. My guess is PTGDS is being increased in human scalp by AR or ERb.
    Quote Originally Posted by InBeforeTheCure View Post
    Yeah, here's the graph that Cotsarelis presented:



    So it steadily rises during anagen then spikes in very late anagen, soon followed by catagen.

    The working hypothesis at Kythera was through AR:



    Either way, certainly downstream of androgens.
    Quote Originally Posted by HairlossAt15 View Post
    Chemical, you say PGD2 does not play the major role, but what about the fact that genetically altered mice which overexpress Ptgs2 phenocopy AGA?

    Transgenic mice overexpressing Ptgs2 in the epidermis phenocopy AGA

    Given the correlation of increased levels of PGD2 with balding scalp in humans and the presumptive inhibitory role of PGD2 on the mouse follicle, we hypothesized that mice with high levels of PGD2 in the skin might develop features of AGA. Because Ptgs2 (cyclooxygenase 2, prostaglandin G/H synthase) is the enzyme upstream to Ptgds, we further hypothesized that mice overexpressing Ptgs2 would have elevated PGD2 levels. Transgenic mice that overexpress Ptgs2 in the epidermis had been developed previously for carcinogenesis studies. The hair follicles in these K14-Ptgs2 transgenic mice were noted to enter catagen prematurely, and these mice reportedly developed alopecia and sebaceous gland enlargement.

    We further analyzed the skin and hair follicles of the K14-Ptgs2 mouse. These mice developed alopecia, which was evident as a decrease in the normal murine pelage coat compared to control (Fig. 5, A and B). By histology, we also detected sebaceous gland hyperplasia as indicated by enlarged sebocytes clustered around the hair follicle (Fig. 5, C and D). The hair follicles in the K14-Ptgs2 mice were miniaturized compared to controls (Fig. 5, C and D), and these follicles bore a marked resemblance to the miniaturized follicles in human bald scalp.

    To determine the prostaglandin content in the alopecic skin of the K14-Ptgs2 mice, we measured prostaglandin levels by HPLC-MS during the anagen phase of the hair follicle cycle. PGE2 was elevated in the K14-Ptgs2 mice compared to age-matched wild-type controls, as was previously shown using immunoassays measuring PGE2 and PGF2α content in biopsied mouse skin. PGD2 was also elevated and was more abundant than PGE2 in both wild-type and K14-Ptgs2 mice. 15-dPGJ2 was elevated in K14-Ptgs2 mice compared to controls and demonstrated the largest fold increase (~14-fold), although baseline values were low (5.7 ng/g tissue) (Fig. 5E). We found low levels (18.4 ng/g tissue) of PGF2α, and an absence of prostacyclin (6k-PGF1α) and thromboxane (TxB2) (Fig. 5E), which are not known to be present in normal skin. Together, the balding phenotype in these mice is likely a result of the overwhelming PGD2 and 15-dPGJ2 inhibitory effects on the hair follicle, despite the presence of PGE2, a known promoter of hair growth.
    Quote Originally Posted by Chemical View Post

    To determine the prostaglandin content in the alopecic skin of the K14-Ptgs2 mice, we measured prostaglandin levels by HPLC-MS during the anagen phase of the hair follicle cycle. PGE2 was elevated in the K14-Ptgs2 mice compared to age-matched wild-type controls, as was previously shown using immunoassays measuring PGE2 and PGF2α content in biopsied mouse skin (14, 24). PGD2 was also elevated and was more abundant than PGE2 in both wild-type and K14-Ptgs2 mice. 15-dPGJ2 was elevated in K14-Ptgs2 mice compared to controls and demonstrated the largest fold increase (~14-fold), although baseline values were low (5.7 ng/g tissue) (Fig. 5E).
    InBeforeTheCure has shown that PGD2 can increase GSK3β, and so it would be incorrect of me to say PGD2 doesnt have a role. I think it has a role but not as significant as the parent mediator of PTGDS: Androgens. Kythera believes PTGDS expression is induced by androgens and recent evidence that PKC activates the PTGDS gene expression (PKC is downstream of Androgens) supports this model. I believe we should be focusing more on Androgen as that way we can eliminate multiple bad pathways with one big swoop.

    Furthermore, I'd encourage you to read the previous page where I mention cotsarelises findings on actual real world PGD2 expression levels in bald scalp, which shows a modest 2.5-4 fold increase. InBeforeTheCure has also brought to light that 50% if AGA individuals are not sensitive to PGD2 further reinforcing my statement that PGD2 is not the only or most significant factor implicating AGA.

    Although the analysis done on mice showed hair phenotype resembling AGA, you can see I've bolded the bit that says "15-dPGJ2 was increased 14 fold" in the Ptgs2 overpressed mice. if you look at figure 2F: http://www.ncbi.nlm.nih.gov/pmc/arti...975/figure/F2/

    You can see that 15-dPGJ2 is only a small fraction of PGD2, so a 14 fold increase in the metabolite would mean a ridiculously high concentration of PGD2 in the mice. Its not at all surprising that suraphysical levels of PGD2 can retard hair growth. But we have to be realistic here, in AGA humans its not that high anyway. If you inject mice with testosterone or DHT, they also show follicle miniturisation exactly like humans. In fact, anything that reduces β-Catenin (antibody or what have you) will result in AGA phenotypes. I hope this has clarified my opinion on PGD2's effects on human hair.

  9. #9
    Member
    Join Date
    May 2014
    Posts
    75

    Default Procyanidin - Apple Polyphenols - PKC - Protein Kinase C

    I've been doing some more reading on PKC and understood a little better how it works with regards to hair growth. PKC caught my attention when I read this study on procyanidins (found in apple skin) increasing hair growth.

    The confusing thing was that PKC can actually inactivate (phopsphorylate) GSK3β and we know GSKβ inhibition increases β-Catenin. So procyanidins mediated PKC inhibition had to be working through a different mechanism.

    Then I found this study:


    Protein-kinase-C-mediated -catenin phosphorylation negatively regulates the Wnt/β-catenin pathway


    A23187 = PKC activator

    A23187-mediated β-catenin degradation requires the β-catenin N-terminus but not GSK-3β activity

    Since the phosphorylation of β-catenin by GSK-3β and its subsequent association with β-TrCP leads to β-catenin degradation, we examined whether A23187-mediated inhibition of CRT requires GSK-3β activity. To this end, HEK293 reporter cells were incubated with A23187 and LiCl, an inhibitor of GSK-3β. As shown in Fig. 2A, A23187 suppressed LiCl-induced CRT. Furthermore, Western blot analysis using anti-β-catenin antibody consistently showed that A23187 reduced the level of β-catenin that accumulated with LiCl treatment (Fig. 2B), indicating that A23187-mediated inhibition of the Wnt/β-catenin pathway is independent of GSK-3β.
    This is quite surprising. β-catenin can be degraded regardless of GSK-3β. This is not good news at all. So even PGE2 and minox's effectiveness will be greatly diminished if PKC is elevated.

    And get this, PGD2 can activate PKC.

    These results strongly suggest that PGD2 stimulates IL-6 synthesis through intracellular Ca2+ mobilization in osteoblasts, and that the PKC activation by PGD2 itself regulates the over-synthesis of IL-6.

    http://www.ncbi.nlm.nih.gov/pubmed/10582659
    We previously showed that PGD2 stimulates the induction of heat shock protein 27 (HSP27) via protein kinase C (PKC)-dependent p38 mitogen-activated protein (MAP) kinase and p44/p42 MAP kinase in osteoblast-like MC3T3-E1 cells.

    http://www.ncbi.nlm.nih.gov/pubmed/15743775
    And PKC can in turn increase PTGDS expression:

    Protein kinase C activates human lipocalin-type prostaglandin D synthase gene expression through de-repression of notch-HES signaling and enhancement of AP-2 beta function in brain-derived TE671 cells.

    Bear in mind these are in different cell types so its not 100% accurate.

    If PGD2 increases PKC then it could mean that in some people the increase in PKC mediated β-catenin degradation may offset its GSK3β inhibiting effects (net yield of no change in β-Catenin). PGD2 also inactivates AKT (InBeforeTheCure) so GSK3β is further increased doubling up on the PKC ¬ β-catenin.

    Another related protein kinase is PKA that has the ability to prevent β-Catenin degradation by occupying the SER675 position that the proteosomes require to carry out the ubiquitinition (study).

    This is why forskolin can boost the effect of procyanidins on hair growth:

    Apples are known to contain the highest concentration of procyanidin b-2, and the polyphenol extract is quite cheap from amazon uk. (Apple polyphenols contain procyanidin). Or alternatively grape seed extract.

  10. #10
    Member
    Join Date
    May 2014
    Posts
    75

    Default Adipogenesis - PDGF

    Quote Originally Posted by solaeg View Post
    I am currently using oleuropein since it seems to have great potential when using it topically, but also internally in terms of body-recomposition and various health related effects (e.g. http://suppversity.blogspot.dk/2012/...tosterone.html and http://www.ergo-log.com/oleuropein-b...anabolism.html)

    As I understand, some of it's fat loss effects comes from being an PPAR Gamma antagonist. Meanwhile, PPAR Gamma agonists stimulates adipogenesis. And a thickening fat layer in the scalp is good for hair (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3992657/ - "In each case, the thickness of the fat layer was decreased by 50% in the regions of hair loss"), while it is otherwise replaced by fibrotic tissue (collagen).
    If I understand it correctly, activation of PPAR Gamma also has anti-fibrotic effects (maybe due to regulating adipogenesis?): http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3733781/

    Considering the above, are there any issues when using Oleuropein topically and/or internally?

    And what is the role of oleuropein and adipogenesis in the scalp?
    "Wnt/β-catenin signaling stimulates adipocyte differentiation in vivo and in vitro": http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3992657/
    On the other hand, Wnt6, Wnt10a and Wnt10b seem to do the opposite:
    http://www.ncbi.nlm.nih.gov/pubmed/21872687
    And I guess we don't want osteoblast in the scalp?

    Is oleuropein activating wnt10b which upregulates wnt/β-catenin signaling which positively regulates adipogenesis in the scalp? Or is it likely to not have an effect on adipogenesis?
    WNT10b inhibits adipogenesis via a mechanism I dont fully understand, along with WNT6 and 10a like you pointed out. You wont have progenitor cells differentiate into osteoblasts in the skin so thats nothing to worry about, but WNT10b will promote the keratinocyte lineage.

    Oleuropein is a known inhibitor of PPARγ, and adipocytes differentiate/mature via PPARγ.

    What happens if the fat layer in scalp reduced? Does the fat layer cause hair regrowth? These studes are excellent and help us understand the underlying mechanics.

    Epidermal Wnt/β-catenin signaling regulates adipocyte differentiation via secretion of adipogenic factors

    Unraveling hair follicle-adipocyte communication

    The adipocyte precursors actually promotes anagen development via PDGF (not to be confused with PGD2).

    In addition to the requirement of adipocytes in the skin for anagen induction, adipocyte lineage cells are sufficient to induce the hair follicle cycle. Transplantation of adipocyte progenitor cells intradermally into the backskin of shaved mice at the extended 3–4 week telogen stage of the hair follicle cycle that occurs around 7 weeks of age resulted in adipocyte graft formation and corresponding precocious hair growth. Anagen was induced in these mice injected with the enriched adipocyte progenitor cells from WT or AZIP, but not with cells of the entire stromal vascular fraction (SVF), supporting that the hair-inducing activity was specific for immature adipocyte lineage cells.

    The mechanism behind this interaction is not completely understood, but PDGF signaling may play a role. PDGFA mRNA is significantly elevated in adipocyte precursor cells, and mice lacking PDGFA show a delay in hair follicle stem cell activation that mirrors the phenotype of Ebf1−/− mice.

    Based on the expression of PDGF ligands by adipocyte lineage cells, the activation of the PDGFR in the DP during anagen, and the ability of PDGF-coated beads to rescue hair cycling defects in Ebf1 null mice, we propose that adipocyte precursor cells secrete PDGF to promote hair growth. Mice lacking PDGF-A display similar hair follicle defects as Ebf1 null mice, namely a lack of anagen entry.
    Only the precursor adipocytes produce PDGF, not the mature differentiated hypertrophic cells.

    How does PDGF do this? look at this:

    Involvement of platelet-derived growth factor receptor-alpha in hair canal formation.

    Hair follicles develop and are maintained by multiple rounds of inductive events involving interactions among various cell types within the follicles and the adjacent mesenchyme. In this study, we established the antagonistic monoclonal antibody APA5 against platelet-derived growth factor (PDGF) receptor-alpha (PDGFR-alpha) and used it to investigate the role of PDGFR-alpha in neonatal skin development. In addition to the dermal mesenchyme, a known site of PDGFR-alpha expression, immunohistologic staining of neonatal skin detected transient expression of PDGFR-alpha in the perinatal epidermis for several days. On the other hand, ligands for PDGFR-alpha were detected in epithelial cells and sebaceous glands of hair follicles. To determine whether this contiguous expression of PDGF and PDGFR-alpha in neonatal skin plays a functional role, we injected APA5 into neonates to block the function of PDGFR-alpha. Consistent with the PDGF/PDGFR-alpha expression in the neonatal skin, two defects were induced by this procedure. First, hair canal formation in the epidermis was severely suppressed. Second, the growth of dermal connective tissues and of hair follicles of pelage hairs was suppressed. These results indicate that PDGF signals are involved in both the epidermis-follicle interaction and the dermal mesenchyme-follicle interaction required for hair canal formation and the growth of the dermal mesenchyme, respectively.
    So the adipocyte precursors (mesenchymal stem cells?) actually signal to the hair follicle canal to prepare for the new anagen cycle. This is the kick that telogen hairs need to actually form the follicle canal before the DPC can start proliferating.

    Effect of IGF-I on Hair Growth Is Related to the Anti-Apoptotic Effect of IGF-I and Up-Regulation of PDGF-A and PDGF-B

    IGF-1 upregulates PDGF expression quite potently which is why IGF-1 is such a powerful mitogenic agent for hair.

    Okay, so what controls adipogenesis fate?

    The growth of the intradermal adipose depot could occur through adipocyte hypertrophy or adipogenesis. While adipocyte hypertrophy involves lipogenesis, adipogenesis requires the proliferation and specification of adipocyte precursor cells into preadipocytes, which exit from the cell cycle and differentiate into mature, lipid-laden adipocytes. Adipogenesis requires the upregulation and transcriptional activity of the nuclear receptor, PPARγ in preadipoctyes
    Mature adipocytes also express signaling molecules that can modulate hair cycling. Intradermal adipocytes express BMP2 maximally in late anagen and early telogen, causing follicles to be refractory to activation cues. Given that BMP signaling blocks anagen induction, these data suggest that adipocyte derived BMPs may promote and maintain follicular stem cell quiescence. Thus, when BMP signals are reduced in the macroenvironment, the hair follicles are open to activation signals, enter into a competent telogen phase and the follicles can re-enter anagen. The contribution of adipocyte derived BMP proteins is unknown since the dermal papillae also express BMP mRNAs.
    Immature adipocytes only help with anagen induction and do not play a huge part in maintenance (Beta Cetenin does that). Mature adipocytes actually secrete factors that inhibit hair growth. This inhibition is probably weak since females hair ridiculously long anagen cycles even though they have a normal scalp fat layer. The adipocytes also have estrogen receptors which could explain their hypertrophy during anagen:

    Delving deeper:

    Analysis of BrdU incorporation within adipocyte precursor cells revealed that prior to anagen ~50% of adipocyte precursor cells are proliferating. However, once anagen was initiated, the percentage of proliferative adipogenic cells was reduced to ~25% (Figure 2C). Thus, adipocyte precursor cells are stimulated to proliferate during late catagen to generate an increased population of adipogenic cells during anagen induction. These data correlate with the timing of de novo adipocyte generation after anagen induction (Figure 1C).

    Thus, these three mouse models with diminished or absent intradermal adipocytes affect different stages of adipogenesis in the skin. The Ebf1 null mouse lacks adipocyte precursor cells suggesting that this mutation acts at the adipocyte precursor cell to block postnatal intradermal adipogenesis. PPARγ antagonists do not block the formation of adipocyte precursor cells in the skin but disrupt the formation of PPARγ+, preadipocytes, resulting in a loss of postnatal intradermal adipogenesis.

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3298746/
    I suspect whats happening here is that once hairs enter anagen they start secreting factors that reduce PPARy signalling, hence the reduction in adipocyte precursors. WNT10b, 10a, and WNT6 could be elevated in anagen hairs, and when the catagen phase kicks in, the adipocyte precursors are free to proliferate, then once the hair follicle is recreated the precurors have done their job. The increase in the fat layer is caused by the growing hair follicles leading to adipocyte hypertrophy:

    During activation of hair growth, the expansion of the intradermal adipocyte layer in the skin doubles the skin’s thickness (Butcher, 1934; Chase et al., 1953; Hansen et al., 1984).
    This means the balding skin is primed for growth, and just needs that kick. The follicles need to be stimulated with WNT/β-catenin to start the adipocyte precursor proliferation and then once the hair canal is formed, you need sustained β-catenin to maintain hair follicle which is a hard job because theres multiple pathways working against you.

Similar Threads

  1. Could anyone elaborate on the science of this?
    By Slam1523 in forum Cutting Edge / Future Treatments
    Replies: 8
    Last Post: 07-20-2023, 09:52 PM
  2. Need some help with the science Docs and you smarties!
    By JayM in forum Men's Hair Loss: Start Your Own Topic
    Replies: 0
    Last Post: 05-23-2015, 08:08 AM
  3. Science + L'Oreal
    By Kiwi in forum Cutting Edge / Future Treatments
    Replies: 1
    Last Post: 06-29-2012, 05:19 PM
  4. Hair Loss Science
    By DepressedByHairLoss in forum Cutting Edge / Future Treatments
    Replies: 13
    Last Post: 03-01-2011, 08:24 PM
  5. LA Science hair serum
    By zxc in forum Hair Loss Treatments
    Replies: 3
    Last Post: 11-07-2010, 06:14 AM

Posting Permissions

  • You may not post new threads
  • You may not post replies
  • You may not post attachments
  • You may not edit your posts

» IAHRS

hair transplant surgeons

» The Bald Truth

» Recent Threads

Which is the best Salesforce institutes in Hyderabad?
Yesterday 09:59 AM
Last Post By palashmim2022
Yesterday 09:59 AM
The Resveratrol microneedling process study - enroll now
04-11-2022 02:38 AM
Last Post By Briam1930
03-16-2024 05:43 AM
What wedding packages are offered in Tbilisi?
01-27-2024 04:13 AM
by Jiromen
Last Post By Henryclark
03-16-2024 03:12 AM
Tour Operator's Treasures in Bangladesh
09-28-2023 06:48 AM
by davidm
Last Post By Briam1930
03-16-2024 12:49 AM