Friday, June 29, 2012

Height increase with Chitosan?

Chitosan is a soluble dietary fiber but it is insoluble in water.  The trouble is getting it from the stomach to the bone.  PCL beads are used for the controlled release of drugs.  Chitosan scaffolds have been used to induce chondrogenesis before. Chitosan is available for sale but can it increase serum levels in the epiphyseal bone marrow is the question: NOW Foods Chitosan 500mg, 240 Caps.

Chondrogenesis of human bone marrow mesenchymal cells by transforming growth factors β1 through cell shape changes on controlled biomaterials.

"The phenotypic responses of human bone marrow mesenchymal cells (hBMSCs) on different ratio of chitosan/polycaprolactone (PCL) blends were investigated in this study. The results showed that hBMSCs existed different morphology on chitosan/PCL blends due to the different adhesion characteristic of cell on neat PCL and neat chitosan. Interestingly, comparing to hBMSCs on neat PCL, hBMSCs aggregated to form spheroid and to express ascendant trend of transforming growth factor β1, collagen type II, collagen type X, and Sox9 mRNA on the chitosan/PCL blended substrates with the decrease of PCL content. To confirm chondrogenesis of hBMSCs with spheroid on test substrates, Alcian Blue and Safranin O staining were used to detect the cartilaginous extracellular matrix (ECM). It revealed hBMSCs with spheroid on neat chitosan and 10 wt % PCL did turn to chondrogenic differentiation and synthesize cartilaginous ECM. Therefore, these findings provided new insights into the role of chitosan/PCL blended material could mediate the endogenous gene expression of hBMSCs to alter the phenotypic behavior through mediating the cell shape."

"The cell–materials interaction is generally considered to be a multistep process involving the adsorption of extracellular matrix (ECM) proteins onto the substrate surface and the recognition between the transmembrane receptors on the cell surface (i.e., integrin) and the cell-binding domain of ECM proteins."

"Focal adhesion kinase is a cytoplasmic tyrosine kinase that is activated on integrin ligating to the ECM at sites of focal adhesion, which results in changes of actin cytoskeleton organization and in the cell spreading, growth, and phenotype"<-Maybe LSJL by inducing Lateral compression and thus alter integrin linking.  Note that lateral compression is much easier to achieve than axial compression.

"[The] gene of transforming growth factor-β1 (TGF-β1) could be upregulated by breaking the cytoskeleton with chemical reagent to make dermal fibroblasts presenting round shape."<-We find out what this chemical reagent is to help us grow taller.

" the TGF-β1 gene expression of cell could be regulated by modulating the cell shape. TGF-β1 was needed for the in vitro chondrogenic differentiation of hBMSCs. We hypothesized that using the chitosan and PCL blended material could alter the gene expression and the phenotypic response of hBMSCs through mediating hBMSCs to present different cell shape on the chitosan and PCL blends."

"On chitosan, hBMSCs exhibited round shape and formed short radial-growth of filopodia. With the increase of PCL content in the blends, hBMSCs presented more flattened shape and spindle-like morphology."<-So this is what we want to happen more rounded shape and more radial growth of filopodia in the stem cells of the epiphyseal bone marrow.

There has to be some way to increase the chitosan content of the bone marrow.

Here's the study that talks about chemical regeants to break up the actin cytoskeleton:

Cytoskeleton Regulates Expression of Genes for Transforming Growth Factor-b1 and Extracellular Matrix Proteins in Dermal Fibroblasts

"Cytoskeleton not only controls cell morphology but also regulates cell growth, migration, differentiation, and gene expression. We have recently reported that reorganization of cytoskeleton induces expression of mRNA for transforming growth factor-beta 1 (TGF-beta 1), collagenase, and tissue inhibitor of metalloproteinase-I (TIMP-I){up in LSJL} in dermal fibroblasts[it's possible that the same chemicals will work on stem cells as well]. In this report we have examined the role of gene transcription in this induction. As judged by nuclear run-on assay, trypsin, EGTA (ethylene glycol-bis (beta-aminoethyl ether) N, N, N', N', tetra-acetic acid), or cytochalasin C (Chs) increased the rate of transcription of the TGF-beta 1 gene by 2.0, 2.7, and 1.6 fold, respectively[So EGTA was most effective], and of the collagenase gene by 5.3, 6.2, and 3.3 fold. The rate of transcription of the TIMP-I gene was increased by trypsin (4.3 fold) or EGTA (3.8 fold) but unaffected by Chs. Cytochalasin induced an increase in the rate of transcription of procollagen I (alpha 1), procollagen I (alpha 2), and fibronectin genes by 1.4, 1.5, and 1.9 fold respectively, while trypsinization or EGTA treatment had no or little effects on these gene. Since transcription of the TGF-beta 1 gene is believed to be largely governed by the activating protein 1 (AP1) complex, we also examined the expression of mRNA for c-fos[LSJL upregulates c-fos] and c-jun protoon-coproteins. Trypsinization induced rapid (within 30 min) and transient expression of c-fos mRNA. A 2.4 fold increase in c-jun mRNA was apparent after 4 hr and persisted for at least 24 hr. Actinomycin D (Act D) suppressed the induction of TGF-beta 1 mRNA by Chs but had less effect on the TGF-beta 1 mRNA in trypsinized cells which had been replated for 4 hr, suggesting that the half life of TGF-beta 1 mRNA is reduced in cells with a disassembled cytoskeleton. Simultaneous treatment with Chs and cycloheximide (Cxm) resulted in a superinduction of TGF-beta 1 mRNA by 88 +/- 23% (n = 4, P < 0.05), which was abrogated by preexposure to Act D. In contrast, the induction of collagenase mRNA by Chs was totally blocked by Cxm, indicating that the Cxm-mediated superinduction is selective and that protein synthesis is required for induction of this mRNA. Our results suggest that the activities of genes for proteins involved in the structure (Type I collagen and fibronectin), turnover (collagenase and TIMP-1) and regulation (TGF-beta 1) of extracellular matrix (ECM), are all governed at least in part by the status of the cytoskeleton."

This study mentioned that chitosan(and GMS) increased serum levels of GH and IGF-1 but didn't directly mention Chitosan serum levels: Effect of dietary supplementation of chitosan and galacto-mannan-oligosaccharide on serum parameters and the insulin-like growth factor-I mRNA expression in early-weaned piglets.

Chitosan is insoluble in water so if it does get past the digestive system and into the blood stream it should stay intact and increase the serum content of chitosan in the epiphyseal bone marrow.

According to Effects of habitual chitosan intake on bone mass, bone-related metabolic markers and duodenum CaBP D9K mRNA in ovariectomized SHRSP rats.,  chitosan can cause bone loss which may favor neo-growth plate formation by making room for it.

So maybe supplementing with chitosan and a glass of water could help you grow taller by encouraging epiphyseal bone marrow stem cells to become a more rounded pro chondrogenic shape.  

Thursday, June 28, 2012

Do you grow when you sleep?

Getting more sleep is one of the most commonly cited ways to grow taller but does it actually help you grow?  Does it only affect growth rate or can it increase adult stature?  Microgravity has been found to be pro-chondrogenic.  When you lay down there's less load on the bone.


Growing pains: are they due to increased growth during recumbency as documented in a lamb model?


"The rate and patterns of longitudinal bone growth are affected by many different local and systemic factors; however, uncompromised growth is usually considered to be smoothly continuous, with predictable accelerations and decelerations over periods of months to years. The authors used implanted microtransducers to document bone growth in immature lambs. Bone length measurements were sampled every 167 seconds for 21 to 25 days. The authors show that at least 90% of bone elongation occurs during recumbency and almost no growth occurs during standing or locomotion[so the more you are recumbant the faster you'll grow but that doesn't mean that your adult height will be larger]. The authors hypothesize that growth may also occur in children during rest or sleep, thus supporting the concept of nocturnal growth and perhaps a relationship to growing pains."

"Within a brief 1- to 3-day interval, children grew 1 cm or more, and this was followed by a much longer period of 10 to 50 days when no demonstrable growth occurred."<-This supports the theory that growth involves a build up and than an explosion where bone is pushed apart.  A build up of hypertrophic chondrocytes followed by apoptosis which pushes the bones apart allowing for new bone to be filled in.

"we found that overall growth consists of a period of rapid elongation with low-amplitude fluctuations. This pattern alternates with high-frequency fluctuations with no or negative growth as measured by microtransducers. The oscillations recorded by both transducers are thought to represent strains at the growth plate and in diaphyseal bone. We interpret these larger oscillations as growth plate compression (negative direction) and elastic recoil and/or growth plate tension (positive direction). Similarly, the oscillations in the diaphyseal transducer signal, while much smaller, are synchronous with oscillations from the growth plate transducer."<-Now just because most growth occurs during recumbancy doesn't mean that other activities can't benefit growth.  LSJL and other activities help with the build up phase before the explosive push that occurs during the recumbant phase.  It makes sense that the push comes during the recumbant phase as there is less force to overcome so the hypertrophic chondrocytes apoptosis' can push the bone apart farther to allow for more bone growth to occur in between.

" including increased growth at rest [may be] due to elevations in growth hormone (GH) and insulin growth factor (IGF-1)"<-this is a confounding variable as to whether recumbancy is the only factor affecting growth.

"when growth plate chondrocytes are stimulated by mechanical strain, the inhibitory loop may prevent bone elongation. It is possible that recumbency leads to decreased strain in the growth plate, with subsequent decreased synthesis of Ihh. Therefore, the Ihh-PTHrP inhibitory pathway is turned off and a cohort of growth plate chondrocytes would proceed to terminal differentiation characterized by hypertrophy, the phase during which most bone growth occurs."<-a mechanism as to how reduced load may trigger hypertrophy and in turn apoptosis.

Here's an article that explains how genes are altered during sleep and height is affected by genes:


A metabolic-transcriptional network links sleep and cellular energetics in the brain.


AMPK may mediate the inhibition of Lin28 activity and Lin28 is pro-height.

"When the AMP/ATP ratio increases due to consumption of ATP (a decrease in cellular “energy charge”), AMPK is phosphorylated by upstream kinases and initiates changes in the cell that slow ATP consumption. This mechanism appears to be at work in the brain in association with sleep; temporal patterns of AMPK phosphorylation provide evidence that this metabolic sensor reacts to changes in sleep/wake states. AMPK phosphorylation in the brain is reduced during times of day when sleep predominates. Enforced wake caused an increase in phosphorylated AMPK (pAMPK)"<-this should be true in non-brain areas like the bone.

NADPH oxidase 2 and 4 are required for chondrogenic differentiation.

"A high rate of ATP production will alter the NAD(P)H:NAD(P) + ratio."  However, the study mentions that sleep deprivation increases oxidative stress thus likely increasing NOX2 and NOX4.  However, Nox2/4 are important for the early stages of chondrogenesis.  Thus, loading, which induces oxidative stress, may be important for the initial stages of chondrogenesis and sleep may be important for later stages of chondrogenesis where there's less oxidative stress.

" GSK3β, like AMPK, is sensitive to cellular metabolic status and enforced wakefulness, and affects sleep. The phosphorylation of GSK3β increases in synaptosomes during enforced wakefulness"

"Overexpression of GSK3β shortens circadian period in cultured mammalian cells while pharmacological inhibition lengthens it."

phosphorylation of GSK3Beta promotes chondrocyte hypertrophy.  GSK3Beta degrades Beta-Catenin. Runx2 expressing cells must have virtually no Beta-Catenin.  This is not consistent with our theory as phosphorylation of GSK3Beta increases during wakefullness and GSK3Beta phosphorylation promotes chondrocyte hypertrophy.  However, the phosphorylation of GSK3Beta was increased in synaptosomes and this may or may not be true for stem cells or chondrocytes.

PPARgamma stimulates adipogenesis and thus less likely to stimulate chondrogenesis.

"PPARs, a family of transcriptional regulatory nuclear receptors, are sensitive to the endogenous sleep-promoting lipids anandamide and oleylethanolamide"

Sleep, ghrelin, leptin and changes in body weight during a 1-year moderate-intensity physical activity intervention.


"We randomly assigned 173 post-menopausal sedentary overweight (body mass index >or=24.0 kg/m(2) and >33% body fat) women aged 50-75 years living in western Washington State to either a facility- and home-based moderate-intensity physical activity intervention or a stretching control group. Fasting plasma ghrelin, leptin, measured height, weight and self-reported sleep were assessed at baseline and 12 months.
There were no consistent cross-sectional patterns between self-reported sleep measures and ghrelin or leptin at baseline. The weight loss differences between exercisers and stretchers were greater for those who slept less at follow-up than at baseline compared to those whose sleep duration did not change (-3.2 kg, 95% confidence interval (CI) -5.8, -0.5). Improvements in sleep quality were associated with significantly greater differences between exercisers and stretchers for ghrelin increases (improved vs same sleep quality: +115 pg/ml, 95% CI +25, +206) and leptin decreases (improved vs worsened sleep quality: -5.7 ng/ml, 95% CI -9.5, -1.5).
There was only limited evidence that changes in sleep duration or quality modified exercise-induced changes in weight, ghrelin or leptin. Moreover, the observed differences were not in the directions hypothesized. Future longitudinal studies including population-based samples using objective measures of sleep and long follow-up may help to clarify these relationships."

<-So sleep enhances exercise increases in Ghrelin and decreases in Leptin.  Remember though that Leptin is growth stimulating.  Ghrelin is expressed in the growth plate.

Now the one most commonly associated with sleep HGH:

Complexity and non-linear description of diurnal cortisol and growth hormone secretory patterns before and after sleep deprivation.


"The circadian secretory profiles of cortisol and growth hormone (hGH) in normal subjects are interrelated. Slight alterations in cortisol secretion are paralleled by similar ones of hGH secretion. Under physiological conditions an inhibitory effect of glucocorticoids on hGH secretion is more potent than a stimulatory one, while in normal young subjects the nychtohemeral cortisol and hGH levels are lower and higher, respectively, post 24 hours total sleep-deprivation, compared to baseline values. The aim of the present work was to further assess the qualitative characteristics of the 24-hour secretory patterns of these two hormones before and after 24 hours total sleep deprivation, by studying their non-linear profiles using fractal analysis.
Cortisol and hGH were measured in 24-hour samples drawn from 10 healthy men (mean age SD: 24 +/- 1 yr, mean BMI SD: 25 +/- 1 kg/m2) before and after 24 hours total sleep deprivation. Twenty-four hour blood sampling was performed serially every 30 min the day before and the day after total sleep deprivation. The 24-hour hormone profiles were analyzed by Fourier spectrum, in order to verify periodicities; the corresponding attractors were drawn and their respective fractal dimensions were calculated using the box counting method.
Diurnal cortisol levels before sleep deprivation gave rise to a fractal attractor with a D0 fractal dimension of 2.65 +/- 0.03, which decreased, post-sleep deprivation, to D0: 2.18 +/- 0.04. Growth hormone before sleep deprivation gave rise to a fractal attractor with a D0 dimension of 1.96 +/- 0.60, which increased to 2.24 +/- 0.60 post-sleep deprivation. These post-sleep deprivation changes of the fractal dimensions of cortisol and hGH, suggest that sleep deprivation leads to a more regular secretory profile of cortisol, while it tends to render hGH secretory profile less regular. Additionally, these changes of the fractal dimensions parallell the previously described quantitative overall changes of these hormones.
The post-sleep deprivation decrease of cortisol fluctuation might reflect the mechanism by which sleep deprivation temporarily improves mood in melancholic depression, a condition associated with hyperactivity of the hypothalamic-pituitary-adrenal axis."

So yes sleep raises HGH and lowers Cortisol.  No genes have yet been found directly linking HGH to height.  There's SOCS2 which affects the HGH/IGF-1 access.  But you'd expect there to be a gene enhancing HGH secretion to increase height if HGH has such a large influence.  However, HGH transgenic mice have been found to be twice as large as their littermates.

So sleep affects load(which is why you shouldn't sleep with ankle weights), AMPK, NADPH, GSK3Beta, PPAR, Ghrelin, Leptin, HGH, and Cortisol.  All of these may have varying effects on height.  However, these hormones may have different effects on stem cells and chondrocytes in different stages of differentiation which is why it is important to cycle recumbancy and activity.  It's possible that a deeper sleep or more recumbancy(weightless sleeping) could have more effects on height.

Saturday, June 23, 2012

Build your Stature by Inhibiting FGF-21?

We know that the periosteum expresses FGF-21.  The periosteum likely plays a key role in height growth.  Here's a study that directly links FGF-21 inhibiting the growth plate:

Fibroblast Growth Factor 21 (FGF21) inhibits chondrocyte function and Growth Hormone (GH) action directly at the growth plate.

"Fibroblast Growth Factor 21 (FGF21) modulates glucose and lipid metabolism during fasting. In addition, previous evidence indicates that increased expression of FGF21 during chronic food restriction is associated with reduced bone growth and Growth Hormone (GH) insensitivity. In light of the inhibitory effects on growth plate chondrogenesis mediated by other FGFs, we hypothesized that FGF21 causes growth inhibition by acting directly at the long bones' growth plate. We first demonstrated the expression of FGF21, FGFR1 and FGFR3 (two receptors known to be activated by FGF21)[we know that FGFR3 is very bad for height growth and can cause dwarfism], and β-klotho (a co-receptor required for the FGF21-mediated receptor binding and activation) in fetal and 3-week old mouse growth plate chondrocytes. We then cultured mouse growth plate chondrocytes in the presence of graded concentrations of rhFGF21 (0.01-10 μg/ml). Higher concentrations of FGF21 (5 and 10 μg/ml) inhibited chondrocyte thymidine incorporation and collagen X mRNA expression. 10 ng/ml GH stimulated chondrocyte thymidine incorporation and collagen X mRNA expression, with both effects being prevented by the addition in the culture medium of FGF21 in a concentration-dependent manner[So chondrocyte thymidine incorporation and collagen X expression might result in more height growth, we know that collagen X expression is a factor in chondrocyte hypertrophy]. In addition, FGF21 reduced GH binding in cultured chondrocytes. In cells transfected with FGFR1 siRNA or ERK 1 siRNA, the antagonistic effects of FGF21 on GH action were all prevented, supporting a specific effect of this growth factor in chondrocytes[So the negative effects of FGF21 on GH action are a result of the increased ERK1 phosphorylation and FGFR1]. Our findings suggest that increased expression of FGF21 during food restriction causes growth attenuation by antagonizing the GH stimulatory effects on chondrogenesis directly at the growth plate. In addition, high concentrations of FGF21 may directly suppress growth plate chondrocyte proliferation and differentiation."

"transgenic mice overexpressing FGF21 are significantly smaller, have reduced tibial length, and display reduced hepatic GH insensitivity when compared to wild-type mice"

FGF21 also increases ERK 1 phosphorylation.

So to grow taller we can either find ways to directly inhibit FGF21 or inhibit FGFR3 and FGFR1.  We already know that CNP inhibits FGFR3.

Lactose may enhance FGF21 signaling:

Dynamics and Distribution of Klothoβ (KLB) and Fibroblast Growth Factor Receptor-1 (FGFR1) in Living Cells Reveal the Fibroblast Growth Factor-21 (FGF21)-induced Receptor Complex.

"FGF21 stimulates FGFR1c activity in cells that co-express Klothoβ (KLB); however, relatively little is known about the interaction of these receptors at the plasma membrane. We measured the dynamics and distribution of fluorescent protein-tagged KLB and FGFR1c in living cells using fluorescence recovery after photobleaching and number and brightness analysis. We confirmed that fluorescent protein-tagged KLB translocates to the plasma membrane and is active when co-expressed with FGFR1c. FGF21-induced signaling was enhanced in cells treated with lactose, a competitive inhibitor of the galectin lattice, suggesting that lattice-binding modulates KLB and/or FGFR1c activity. Fluorescence recovery after photobleaching analysis consistently revealed that lactose treatment increased KLB mobility at the plasma membrane, but did not affect the mobility of FGFR1c. The association of endogenous KLB with the galectin lattice was also confirmed by co-immunoprecipitation with galectin-3. KLB mobility increased when co-expressed with FGFR1c, suggesting that the two receptors form a heterocomplex independent of the galectin lattice. Number and brightness analysis revealed that KLB and FGFR1c behave as monomers and dimers at the plasma membrane, respectively. Co-expression resulted in monomeric expression of KLB and FGFR1c consistent with formation of a 1:1 heterocomplex. Subsequent addition of FGF21 induced FGFR1 dimerization without changing KLB aggregate size, suggesting formation of a 1:2 KLB-FGFR1c signaling complex. Overall, these data suggest that KLB and FGFR1 form a 1:1 heterocomplex independent of the galectin lattice that transitions to a 1:2 complex upon the addition of FGF21."

Now milk and diary have a lot of beneficial effects on height growth.  Maybe lactose free milk will give the benefits of milk without inhibiting galectin lattice.  Lactoferrin can also be taken directly.

There are prescription FGFR inhibitor drugs but you only want to inhibit FGFR1 and FGFR3 not FGFR2 and FGFR4.


Bone Marrow Mesenchymal Stem Cells: Fat On and Blast Off by FGF21.

"activation of the Wnt/β-catenin signaling pathway can stimulate BMMSCs to differentiate into osteoblast but inhibit adipogenesis and chondrogenesis"

"[fgf21] is a powerful regulator of glucose and lipid metabolism. Physiologically, FGF21 expression is induced both in the liver by prolonged fasting via PPARα activation and in the white adipose tissue by feeding via PPARγ activation. Pharmacologically, administration of recombinant FGF21 to diabetic mice and rhesus monkeys strongly enhances insulin sensitivity, [and] decreases plasma glucose and triglyceride"

"genetic loss-of-function as in the FGF21 knockout mice leads to a higher bone mass with less marrow adipocytes but more osteoblasts and accelerated bone formation."

"FGF21 forms a feed-forward loop with PPARγ: ligand activation of PPARγ increases the expression of FGF21 and its co-receptor β-Klotho in BMMSCs; in turn, FGF21 enhances PPARγ activity by attenuating its inhibitory sumoylation"

Friday, June 22, 2012

Height Growing with Calciumfluor?

Recently, someone emailed me about FMS*calciumfluor(ultra loses of Calcium Fluoride associated with Calcium Monophosphate) as a potential height increasing supplement. Like: Calcium Fluoride 6X 500 Tablets  Calciumfluor does stimulate p42 MAPK which stimulates chondrogenesis.

Differential regulation of chondrogenic differentiation by the serotonin2B receptor and retinoic acid in the embryonic mouse hindlimb.

"Retinoic acid (RA) synthesizing and metabolizing enzymes are coordinately expressed with serotonin 2B (5-HT2B)[we've learned about Serotonin possibly having a height increasing before] receptors at sites of epithelial-mesenchymal (E-M) interaction in the mouse embryo. The promoter of the 5-HT2B receptor contains potential RA response element (RAREs) as well as an AP-2 site. Because both retinoid and serotonergic signaling have been implicated in the regulation of chondrogenic differentiation, the present study investigated whether these signals may work together to regulate this morphogenetic process in hindlimb bud micromass cultures. Results indicate that 5-HT promotes [35S]sulfate incorporation (chondrogenic differentiation) by activation of 5-HT2B receptors, which use the mitogen activated protein kinase (p42 MAPK) signal transduction pathway, whereas RA dose-dependently inhibits sulfate incorporation and promotes expression of RARbeta, which could lead to inhibition of p38 MAPK. No evidence was found to support the possibility that RA negatively regulates expression of 5-HT2B receptors. Taken together, these results suggest that 5-HT and RA may act as opposing signals to regulate chondrogenic differentiation in the developing hindlimb, possibly mediated by different MAPK signal transduction pathways."

"5-HT receptor antagonists have been shown to differentially down-regulate cartilage proteoglycan core protein, a marker of cartilage matrix. 5-HT exerts dose-dependent stimulatory effects on levels of insulin-like growth factor-1 (IGF-1), and inhibitory effects on cell proliferation, mediated by 5-HT receptors that activate the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. Activation of 5-HT receptors that either negatively regulate cAMP or activate the Gq-coupled 5-HT2B receptor, promotes cell proliferation in these cultures."

"Serotonin (5-HT) promotes chondrogenic differentiation in hindlimb micromass cultures." "Epithelial 5-HT uptake sites coincide with areas of chondrogenic differentiation in mesenchyme"

"Serum-free hindlimb MMCs were treated with different doses of 5-HT (10−6 M, 10−8 M), together with nialamide (a monoamine oxidase inhibitor) and L-cysteine (an antioxidant), to prevent degradation of 5-HT. Both doses of 5-HT significantly increased [35S]sulfate incorporation, indicative of maturation of cartilage matrix" "IGF-1 significantly promoted [35S]sulfate incorporation"<-So maybe 5-HT and IGF-1 stimulate chondrogenesis by both stimulating sulfate incorporation.

"various 5-HT receptor agonists, including 8-OH-DPAT (5-HT1A) and SC53116 (5-HT4), increase levels of IGF-1, which itself can promote chondrogenesis"

"Activation of the p42/44 MAPK pathway is involved in the positive regulation of chondrogenic differentiation by 5-HT2B receptors in hindlimb cultures."

"RA[Vitamin A] negatively regulates chondrogenic differentiation, without promoting cell death or decreasing 
cell proliferation"

"The 5-HT2B receptor activates phospholipase C beta (PLC-β)[LSJL upregulates PLC], causing hydrolysis of phosphatidylinositol bisphosphate (PIP2) to generate diacylglycerol (DAG) and inositol trisphosphate (IP3). DAG then activates protein kinase C (PKC). In tumor cells and cell lines expressing the 5-HT2B receptor, activation of this receptor causes PKC to activate the p42/p44 MAPK pathway by means of p21 ras"

Serotonin increases chondrogenesis via the p42/p44 MAPK pathway so therefore maybe the p42/p44 pathway can increase chondrogenesis.

FMS*Calciumfluor specifically increases mRNA levels and induces signaling via MAPK 42,44 and not FAK in differentiating rat osteoblasts.

"The continuous exposure of differentiating rat osteogenic cells (ROB) to FMS*[The homeopathic compound of resonance FMS*Calciumfluor] modulates the level of expression of mRNAs for 7 of the 8 osteogenic markers tested. Alkaline phosphatase (AP), osteocalcin (OC), metalloproteinases (MMP-2 and -14), procollagenase C (BMP-1), biglycan (BG) and integrin 1 are expressed at higher levels in FMS*-treated osteoblasts than in control cultures. MMP-2 and -14 mRNA are not down-modulated at mineralization. Also, the pattern of expression induced by FMS* for some of these genes (BMP-1, BG and integrin 1) is changed, but collagen type I (Coll I) mRNA levels are not affected by treatment with FMS*. This suggests that FMS* modulates mRNA levels and that this is not generalized, but gene(s) specific. We also report that exposure to FMS* rapidly and transiently induces activation of mitogen-activated protein kinases (MAPKs) 42,44 in populations of early osteoblasts[so it may activate it in stem cells as well]but not in pre-osteoblasts, with a cell differentiation stage-dependent and pertussis toxin (PTX)-sensitive response. Subsequent to FMS* MAPK signaling activation, an increase in AP and MMP-14 mRNA is detected, which is also inhibited by PTX, suggesting that FMS* activation of MAPK signaling could be an early event required for the induction of these genes. Exposure to FMS* does not cause changes in the activity of p125 (FAK)-mediated signaling."

Here's an article that links Fluoride to Serotonin:

Phosphorylation of serine 526 is required for MEKK3 activity, and association with 14-3-3 blocks dephosphorylation.

"MAPK/ERK kinase kinase 3 (MEKK3) is a mitogen-activated protein kinase kinase kinase (MAP3K) that functions upstream of the MAP kinases and IkappaB kinase. Phosphorylation is believed to be a critical component for MEKK3-dependent signal transduction, but little is known about the phosphorylation sites of this MAP3K. To address this question, point mutations were introduced in the activation loop (T-loop), substituting alanine for serine or threonine, and the mutants were transfected into HEK293 Epstein-Barr virus nuclear antigen cells. MEKK3-dependent activation of an NF-kappaB reporter gene as well as ERK, JNK, and p38 MAP kinases correlated with a requirement for serine at position 526. Constitutively active mutants of MEKK3, consisting of S526D and S526E, were capable of activating a NF-kappaB luciferase reporter gene as well as ERK and MEK, suggesting that a negative charge at Ser526 was necessary for MEKK3 activity[phosphorylation adds a negative charge] and implicating Ser526 as a phosphorylation site[Ser526 activates MAP3K pathway]. An antibody was developed that specifically recognized phospho-Ser526 of MEKK3 but did not recognize the S526A point mutant. The catalytically inactive (K391M) mutant of MEKK3 was not phosphorylated at Ser526, indicating that phosphorylation of Ser526 occurs via autophosphorylation. Endogenous MEKK3 was phosphorylated on Ser526 in response to osmotic stress[LSJL induces osmotic stress]. In addition, phosphorylation of Ser526 was required for MKK6 phosphorylation in vitro, whereas dephosphorylation of Ser526 was mediated by protein phosphatase 2A and sensitive to okadaic acid and sodium fluoride. Finally, the association between MEKK3 and 14-3-3 was dependent on Ser526 and prevented dephosphorylation of Ser526. In summary, Ser526 of MEKK3 is an autophosphorylation site within the T-loop that is regulated by PP2A and 14-3-3 proteins."

So Fluoride can dephoshorylate Ser526.
"the presence of PP2A inhibitors such as okadaic acid or sodium fluoride in cell extracts containing transfected MEKK3 prevented dephosphorylation of Ser526"<-Fluoride prevents dephosphorylation of Ser526 and do keeps the MAPK pathway running.

Here's a study that shows Fluoride can alter chondrogenesis:

[Evaluation of the repair process in mechanically injured rat bone stimulated by sodium fluoride with non-toxic doses].

"The influence of sodium fluoride on the course of repair process in the mechanically injured rat bone was studied. Thirty six male Wistar rats aged 5 months, weighing 460-540 g were investigated. The animals lived under standard conditions and were fed ad libidum with the standard LSM food including 0.7 mg/kg of fluorine on the average. The animals randomly divided into 3 groups that comprised study and control groups, 6 rats each. The rats in the first group were given water with 20 mg (1.05 mmol) of sodium fluoride per kg of body weight for 24 h over a period of 2 weeks--group Ia. In the second group--IIa--animals were given water with sodium fluoride at a dose of 1.5 mmol/kg b.w./24 h for a period of 4 weeks. In the third group--IIIa--the animals were given sodium fluoride in a dose of 1.5 mmol/kg b.w./24 h for a period of 6 weeks. The rats from the control groups I, II and III were given water without sodium fluoride for the period of 2, 4 and 6 weeks, respectively. At the beginning of the experiment a hole was drilled in both femoral bones in rat under barbiturate anaesthesia. According to the protocol the rats underwent ether euthanasia after 2, 4 and 6 weeks after surgery and the following samples were collected: blood from the heart for biochemical studies and both femoral bones for biochemical and histological studies. The following parameters were evaluated in blood serum: fluorine, calcium, magnesium contents, serum concentrations of urea, creatinine, bilirubin and activity levels of enzymes: aspartate aminotransferase, alanine aminotransferase, cholinesterase, base phosphatase. Fluorine, calcium magnesium and zinc contents were estimated in bone samples. The concentration of fluorine ions in animal serum after 2, 4 and 6 weeks of experiment increased significantly as compared with the corresponding controls. The highest fluorine concentrations were observed in serum of rats supplemented with NaF for 6 weeks. The fluorine concentrations in the bone tissue and fresh and dried granulation tissues in all studied groups also revealed statistically significant increase as compared to the controls. The rats fed with sodium fluoride for the period of 6 weeks revealed statistically significant increase of serum magnesium concentration as compared to the remaining study groups. Bone magnesium concentrations in animals fed with NaF for the period of 2 and 6 weeks were higher as compared to the corresponding control groups, with the highest differences observed after 6 weeks of experiment. Animals fed with sodium fluoride for the period of 6 weeks revealed increased serum calcium concentrations as compared to the study groups after 2 and 4 weeks of experiment. Similar results were achieved in bone tissue samples. The use of sodium fluoride led to accelerated chondrogenesis process in the area of insufficiently perfused bone."

So Calcium Fluoride may increase height by increasing chondrogenesis by stimulating the p42/p44 MAPK pathway.  However, most of the studies on fluoride have used sodium fluoride and not calcium fluoride.  Most of the studies have also tested for osteogenesis and not chondrogenesis.

Thursday, June 21, 2012

Inflammation to Grow Tall?

We've learned that HMGA2 is a vital gene for height growth.  That Lin28 can increase HMGA2 by inhibiting Let-7.  However, barring a way to directly stimulate Lin28 or Myc, let-7 inhibition mainly occurs through inflammation driven NF-kappaB pathway.  NF-kappaB promotes stemness but it also causes apoptosis usually resulting in no net change in growth.  Although apoptosis may be involved in height growth.  However, in adult growth plates where there is no cartilage in the bone all the apoptosis will occur in bone cells(of course there is cartilage in the knee joint).  The NF-kappaB pathway no encourages an increase in stemness allowing for chondrogenic differentiation.  So you're trading out bone cells for potential chondrogenic cells.

This may mean that you might want to purposefully induce inflammation in the bone with LSJL in bones that do not have cartilage in them.  The NF-kappaB pathway was found to be crucial to induce chondrogenesis but you still may want to inhibit other inflammatory cytokines like IL-1 and TNF-alpha.  Shear strain(LSJL induces shear strain) has been found to induce IL-6 in chondrocytes, although we don't know if this will occur in bone or stem cells.

Obesity may induce inflammation:

Metabolomic profiling reveals mitochondrial-derived lipid biomarkers that drive obesity-associated inflammation.

"[We] compare traditional lard-based high fat diets (HFD) to "Cafeteria diets" (CAF) consisting of nutrient poor human junk food. [There's a] rapid and severe obesogenic and inflammatory consequences of CAF compared to HFD including rapid weight gain, markers of Metabolic Syndrome, multi-tissue lipid accumulation, and dramatic inflammation. We profile serum, muscle, and white adipose from rats fed CAF, HFD, or standard control diets. Clusters of fatty acids and acylcarnitines [were elevated]. These increases in metabolites were associated with systemic mitochondrial dysfunction that paralleled weight gain, physiologic measures of Metabolic Syndrome, and tissue inflammation in CAF-fed rats. [There were] strong correlations between elevated markers of inflammation in CAF-fed animals, measured as crown like structures in adipose, and specifically the pro-inflammatory saturated fatty acids and oxidation intermediates laurate and lauroyl carnitine. Treatment of bone marrow-derived macrophages with lauroyl carnitine polarized macrophages towards the M1 pro-inflammatory phenotype through downregulation of AMPK and secretion of pro-inflammatory cytokines. Compared to a traditional HFD model, the CAF diet provides a robust model for diet-induced human obesity, which models Metabolic Syndrome-related mitochondrial dysfunction in serum, muscle, and adipose, along with pro-inflammatory metabolite alterations."

So high fat and saturated fat diets may increase inflammation.

"Lauroyl carnitine can activate NFΚB signaling"  Here's L-Carnitine: I'm not sure how it compares to Lauroyl L-carnitine NOW Foods L- Carnitine Tartrate 1000mg, 100 Tablets.

Here's a study showing how heterotopic ossification is modified by inflammation, we don't want heterotopic ossification we want chondrogenesis within the bone(endochondral ossification) but if heterotopic ossification can occur so too should endochondral ossification:

Heterotopic Ossification Following Burn Injury: The Role of Stem Cells.

"Heterotopic ossification (HO) [is] the abnormal development of bone tissue in soft-tissue locations. HO is highly associated with burn injury{can we mimic some of the molecules induced by bone injury within the bone?}."

"a 60% incidence [of heterotopic ossification] is observed in patients with severe burns."<-so induce the same factors caused by severe burns in the bone marrow?

"One example of a primary cause of HO is the genetic disorder fibrodysplasia ossificans progressiva (FOP), in which ectopic ossification progresses episodically and in response to minor trauma beginning in childhood"<-We need to study this disorder because it does induce endochondral ossification.  Keep inducing mini-growth plates in the bone and you'll grow tall forever.

"HO lesions from subjects with FOP were found to stain strongly for endothelial (angiopoietin-1 receptor [TIE-2] and von Willebrand factor) and osteogenic (osteocalcin) markers"

"Endothelial cells transdifferentiate into the mesenchymal stem cells (MSCs) that subsequently form ectopic bone in a process called endothelial-to-mesenchymal transition (EndMT)."<-This is something we don't want to happen.  We want to use the mesenchymal cells already in the bone marrow.

"BMP-2 producing fibroblasts caused both the recruitment of mesenchymal cells and the upregulation of the peroxisome proliferatoractivated receptor  γ pathway. Upregulation of the peroxisome proliferator-activated receptor γ pathway leads to the differentiation of the recruited mesenchymal cells to brown adipocytes. The brown adipocytes then create the hypoxic microenvironment necessary for endochondral ossification to occur by converting nutrients and oxygen into heat through the uncoupling of oxidative phosphorylation."<-So fat in bone can create hypoxic environment for chondrogenic differentiation.

Burn injury involves both hypoxia and inflammation which is likely which it can induce endochondral ossification.  Safer ways to induce these things should be used.

Long-term aerobic exercise reduces inflammation and likely makes cells more resistance to hypoxia.

Here's a study that shows inflammation can be induced by prolonged exercise:

Effect of inflammation induced by prolonged exercise on circulating erythroid progenitors and markers of erythropoiesis.



"Exercise in humans augments the mobilization of circulating hematopoietic progenitor cells (CD34(+)) from the bone marrow. We investigated the effect of inflammation on erythroid marrow activity by mobilization of erythroid progenitor cells (EPs) along with soluble markers of erythropoiesis.
Ten healthy athletes who participated in an ultradistance foot race participated in the study. Peripheral blood mononuclear cells were isolated, before (phase I), at the end (phase II), and at 48 h post-race (phase III). EPs were detected as burst colony forming units (BFU-e) and colonies were scored at day 14. Markers of inflammation (C-reactive protein, serum amyloid-A, interleukin-6, ferritin and S100B) and bone marrow activity (erythropoietin, soluble transferrin receptor and lipocalin-2) were assessed.
An approximately three-fold decrease in BFU-e number was observed at phase II. sTfR concentrations were also decreased at phase II and remained decreased at phase III. However, EPO and lipocalin-2 concentrations reached a maximum value at phase II, with a tendency to decrease at phase III.
Exercise-induced inflammation modulates bone marrow homeostasis leading to an increase in leukocyte turnover and a decrease in erythroid compartment. It appears that lipocalin-2 is the main factor that regulates the production and mobilization of EPs."

IL-6 levels were huge post race but returned to normal after 48 hours.

"The increase in progenitor cells [is] a consequence of mobilization processes occurring in the bone marrow after short supra-maximal exercise, whereas a longer time period leads to a reduction in the amount of cells due to inflammation"

However, there have been no reports of marathon runners growing taller.  Prolonged running should induce both hypoxia and inflammation in the bone marrow.  Running may not induce hydrostatic pressure and there may not be release of TGF-Beta1 to induce chondrogenic differentiation.


"In healthy amateur runners, we collected venous blood before, at the end of, and the day after a marathon race, and before and at the end of a 1.5-km field test, and measured hemopoietic and angiogenetic progenitors, as well as plasma or serum concentrations of several cytokines/growth factors. After the marathon, CD34(+) cells were unchanged, [but] decreased number of colonies for both erythropoietic (BFU-E) and granulocyte-monocyte (CFU-GM) series, returning to baseline the morning post-race. CD34(+) cells[bone marrow derived progenitors], BFU-E, and CFU-GM increased after the field test. Angiogenetic progenitors, assessed as CD34(+)KDR(+) and CD133(+)VE-cadherin(+) cells or as adherent cells in culture expressing endothelial markers, increased after both endurance and maximal exercise but showed a different pattern between protocols. Interleukin-6 increased more after the marathon than after the field test, whereas hepatocyte growth factor and stem cell factor increased similarly in both protocols. Plasma levels of angiopoietin (Ang) 1 and 2 increased after both types of exercise, whereas the Ang-1-to-Ang-2 ratio or vascular endothelial growth factor-A were little affected. Circulating hemopoietic progenitors may be utilized in peripheral tissues during prolonged endurance exercise[so the stem cells are being used up rather than differentiating into chondrocytes to grow taller]. Endothelial progenitor mobilization after exercise in healthy trained subjects appears modulated by the type of exercise. Exercise-induced increase in growth factors suggests a physiological trophic effect of exercise on the bone marrow."

So we're not sure yet how to apply inflammation to grow taller.  Slightly longer durations of LSJL may induce a little inflammation and stimulate the NF-kappaB pathway.

According to Pro-Inflammatory Cytokines Produced by Growth Plate Chondrocytes May Act Locally to Modulate Longitudinal Bone Growth, inflammatory cytokines are produced endogenously by the growth plates and inhibiting these cytokines was shown to increase growth.  "The combined treatment with anakinra plus etanercept (500 + 500 µg/ml) resulted in a growth which was 20.3 ± 1.9% above control bones".  "when metatarsal bones were treated with the combination of anakinra plus etanercept the hypertrophic zone height was significantly increased by 43% and the hypertrophic zone area increased by 69%."  Note those two drugs inhibit IL-1Beta and TNF-alpha without inhibiting IL-6 which is likely why inhibiting those compounds have beneficial effects.  As the body can just use IL-6 for some of the beneficial effects of inflammatory cytokines with less bad effects.

Tuesday, June 19, 2012

Grow taller by manipulating HMGA2?

HMGA2 is a key gene modulating human height with it's expression and it's inhibitor let-7(CDK6 gene) observed in several studies as modulating stature.

HMGA2: A pituitary tumour subtype-specific oncogene?

"The Hmga2 gene has a critical role in the control of body growth and adipocytic cell proliferation and differentiation. Hmga2-null mice showed a pygmy phenotype with a decreased body size of 20% in heterozygous and 60% in homozygous mice, and a drastic reduction of the fat tissue, while transgenic mice expressing a truncated Hmga2 without the 3′ UTR demonstrate gigantism. alleles of SNPs linked to HMGA2 have been found associated with human height, and a germline chromosomal inversion that produces a truncated HMGA2{up in LSJL} gene was identified in a boy with severe overgrowth.
HMGA2-linked SNPs most strongly associated with height lie in the HMGA2 3′ UTR, which is bound by the microRNA (miR) Let-7. The height-influencing genetic event linked to these SNPs may possibly influence Let-7 binding and its consequent downregulation of HMGA2 protein expression."



Full-size image (38 K)
Full-size image (49 K)
Ignore the pituitary transformation part of this but you can see how HMGA2 influences the cell cycle and this may be how HMGA2 alters height growth.

In vivo modulation of HMGA2 expression.

"A phenotypic analysis performed on homozygous mutants at the pygmy locus showed that the adults are 40% of the size of wild-type litter-mates whereas adult heterozygotes are 80% of the size of wild-type litter-mates."

"Hmga2 negatively regulates p16Ink4a and p19Arf"

"HMGA2 expression is not detected in normal adult tissues, low level of HMGA1 expression has been observed in adult tissues of both humans and mice"

Inhibiting let7 may inhibit its binding to HMGA2 allowing it to be expressed in adult tissues.

"targeted knock-out mouse that lacked exons 2 and 3 of Hmga2 [is] referred to as the Hmga2 null mouse (Hmga2−/−). Hmga2−/− mice had the same phenotype that was observed in the pygmy mice and the growth retardation in these mice resulted from a decrease in mesenchymal cell proliferation"

Lin28 is an inhibitor of let-7 so increasing levels of that may allow for HMGA2 expression.

Metformin may antagonize Lin28 and/or Lin28B activity, thereby boosting let-7 levels and antagonizing cancer progression.

"The let-7 family of microRNAs has cancer suppressor activity. Markedly reduced levels of let-7 are largely responsible for cancer stemness.  Metformin opposes cancer cell stemness [and boosts] let-7a levels in cancer stem cells. Metformin may reflect AMPK-mediated{AMPK Beta-2 subchain is downregulated by LSJL} inhibition of the expression or activity of Lin28/Lin28A, proteins which act post-transcriptionally to decrease the levels of all let-7 family members[don't take metformin if you want to grow taller]. The transcription of Lin28B{upregulated by LSJL} is promoted by NF-kappaB and by Myc[stimulating both NF-kappaB and Myc are options]].  [Antagonists for] NF-kappaB or Myc activity [include] salsalate, antioxidants, tyrosine kinase and cox-2 inhibitors[thus you may want to increase tyrosine kinase and COX-2 to grow taller], ribavirin, vitamin D, gamma-secretase inhibitors (when available), and parenteral curcumin may have some utility in this regard."

"The PI3K-Akt-GSK3β pathway downstream from most tyrosine kinase receptors can stimulate Myc transcription by promoting nuclear localization of beta-catenin, and also boost EMT by blocking the GSK3β-mediated nuclear export and proteasomal degradation of Snail. And mTOR activation by growth factors (opposable by metformin) can boost the translation of Myc mRNA"<-methods to increase Myc

"Myc mRNA is one of those so-called “weak” mRNAs whose translation can be boosted by interaction with eIF4E. mTOR activity amplifies the availability of free eIF4E by phosphorylating and thereby disabling 4EBP-1, which functions to bind and sequester eIF4E."<-so other activities that increase free eIF4E may boost height as well.

"STAT3 [induces] Myc"

"Notch1 directly promotes transcription of Myc"

"let-7 [sustains] its own expression by antagonizing Lin28, Ras, and Myc translation"<-So we have to find creative ways to inhibit let-7

"Let-7 expression can be globally but specifically reduced via increased activity of Lin28/Lin28B"

let-7/HMGA2/Lin28 are way more important to research than CNP.  Several genes in height growth gene association studies are related to these.  Find homeopathic remedies or supplements that decrease let-7 or increase HMGA2/Lin28.

This page claims that it's Astragalus increases Lin28 levels and that it's Astragalus has been specifically engineered to increase Lin28(I couldn't find any studies on Astragalus and Lin28).  The site does not seem legit though.

This scientist explains that basketball players may be lin28 phenotypes.

TTP is a target to increase Lin28/decrease let-7/increase HMGA2.

Ectopic over-expression of tristetraprolin in human cancer cells promotes biogenesis of let-7 by down-regulation of Lin28.

"Tristetraprolin (TTP) is a AU-rich element (ARE) binding protein and exhibits suppressive effects on cell growth through down-regulation of ARE-containing oncogenes. let-7 microRNA has emerged as a significant factor in [cell growth] suppression{anything that causes cell growth makes cancer grow faster but it doesn't cause cancer}. Both TTP and let-7 are often repressed in human cancers, thereby promoting oncogenesis by derepressing their target genes. TTP promotes an increase in expression of mature let-7{so we can decrease TTP to decrease Let-7}, which leads to the inhibition of let-7 target gene CDC34 expression and suppresses cell growth. [TTP-mediates] inhibition of Lin28. Lin28 mRNA contains ARE within its 3'-UTR and TTP enhances the decay of Lin28 mRNA through binding to its 3'-UTR. TTP-mediated down-regulation of Lin28 plays a key role in let-7 miRNA biogenesis."

And this study says MAPK represses TTP:


"mRNA turnover is a critical step in the control of gene expression. In mammalian cells, a subset of mRNAs regulated at the level of mRNA turnover contain destabilizing AU-rich elements (AREs) in their 3' untranslated regions. These transcripts are bound by a suite of ARE-binding proteins (AUBPs) that receive information from cell signaling events to modulate rates of ARE mRNA decay. A key destabilizing AUBP, tristetraprolin (TTP), is repressed by the p38 mitogen-activated protein kinase (MAPK)-activated kinase MK2 due to the inability of phospho-TTP to recruit deadenylases to target mRNAs. TTP is tightly associated with cytoplasmic deadenylases and promotes rapid deadenylation of target mRNAs both in vitro and in cells. TTP can direct the deadenylation of substrate mRNAs when tethered to a heterologous mRNA, yet its ability to do so is inhibited upon phosphorylation by MK2. Phospho-TTP is not impaired in mRNA binding but does fail to recruit the major cytoplasmic deadenylases. phosphorylation of TTP by MK2 primarily affects mRNA decay downstream of RNA binding by preventing recruitment of the deadenylation machinery. TTP may remain poised to rapidly reactivate deadenylation of bound transcripts to downregulate gene expression once the p38 MAPK pathway is deactivated."

"Phosphorylation of TTP by the p38-activated kinase MK2 promotes 14-3-3 association and inhibits the ability of TTP to trigger the deadenylation of tethered mRNA in cells by preventing the recruitment of cytoplasmic deadenylases. In contrast, phosphorylation does not affect the ability of TTP to bind mRNA. "<-so it's possible that MAPK doesn't impact the ability of TTP to stimulate let-7.

I shot an email to a writer of the study but Lin28 is a protein not mRNA.

According to RNA-binding protein L1TD1 interacts with LIN28 via RNA and is required for human embryonic stem cell self-renewal and cancer cell proliferation., L1TD1 could increase the effect of Lin28.


"A transient inflammatory signal can initiate an epigenetic switch from nontransformed to cancer cells via a positive feedback loop involving NF-kappaB, Lin28{Lin28B is up in LSJL}, let-7, and IL-6{up in LSJL}. We identify differentially regulated microRNAs important for this switch and putative transcription factor binding sites in their promoters. STAT3, a transcription factor activated by IL-6, directly activates miR-21 and miR-181b-1. Transient expression of either microRNA induces the epigenetic switch. MiR-21 and miR-181b-1, respectively, inhibit PTEN and CYLD{down in LSJL} tumor suppressors, leading to increased NF-kappaB activity required to maintain the transformed state. STAT3-mediated regulatory circuits are required for [transformed states] in diverse cell lines. STAT3 is not only a downstream target of IL-6 but, with miR-21, miR-181b-1, PTEN, and CYLD, is part of the positive feedback loop that underlies the epigenetic switch that links inflammation to cancer."

So we have to study this loop to see if inflammation can induce Lin28.

"Lin28 is a direct target of NF-κB"->So NF-kappaB may actually increase height growth as evidenced by the studies on proepithelin.

"The transcription factor NF-κB [may be] required for inhibition of the Let-7 microRNA family but this regulation is indirect and involves Lin28 as an intermediary protein"<-so you may be able to get around NF-kappaB by directly stimulating Lin28 in other ways.


"Src activation triggers an inflammatory response mediated by NF-kappaB that directly activates Lin28 transcription and rapidly reduces let-7 microRNA levels. Let-7 directly inhibits IL6 expression, resulting in higher levels of IL6 than achieved by NF-kappaB activation. IL6-mediated activation of the STAT3 transcription factor is necessary for transformation, and IL6 activates NF-kappaB, thereby completing a positive feedback loop."

"Phosphorylation of STAT3, a downstream target of IL6, is inhibited by the addition of let-7a or by inhibition of Lin28B"<-phosphorylation of STAT3 may be a way to increase height.

"IL6 treatment inhibits let-7a microRNA expression in a manner that depends upon NF-κB"

More on TTP:

Novel phosphorylation-dependent ubiquitination of tristetraprolin by mitogen-activated protein kinase/extracellular signal-regulated kinase kinase kinase 1 (MEKK1) and tumor necrosis factor receptor-associated factor 2 (TRAF2).

"TNF-receptor 1 engagement concomitantly activates NF-κB and JNK signaling. The correctly timed activation of these pathways is the key to account for the balance between NF-κB-mediated cell survival and cell death, the latter fostered by prolonged JNK activation. Tristetraprolin (TTP), initially described as an mRNA destabilizing protein, acts as negative feedback regulator of the inflammatory response: it destabilizes cytokine-mRNAs but also acts as an NF-κB inhibitor by interfering with the p65/RelA nuclear import pathway. TTP contributes to the NF-κB/JNK balance. MAP 3-kinase MEKK1 [is a] TTP kinase that, together with the TNF receptor-associated factor 2 (TRAF2), constitutes not only a main determinate of the NF-κB-JNK cross-talk but also facilitates "TTP hypermodification": MEKK1 triggers TTP phosphorylation as prerequisite for its Lys-63-linked, TRAF2-mediated ubiquitination[So MEKK1 is another target for height increase]. TTP no longer affects NF-κB activity but promotes the activation of JNK.  Upon TNFα induction, TTP transits a hypo- to hypermodified state, thereby contributing to the molecular regulation of NF-κB versus JNK signaling cascades."

So we don't want MEKK1 as we want TTP to go down the NF-kappaB cascade.  So inhibiting MEKK1 could help increase height.

"The MAPK p38α induces TTP phosphorylation, traceable by a “molecular weight shift” of the protein in Western blots, inactivating TTP mRNA binding and degrading ability, but in contrast, p38α did not block TTP function toward NF-κB"<-Thus why we want MAPK38 to be activated and not MEKK1.

"coexpression of MEKK1, which has not been described as TTP kinase yet, could even counteract TTP inhibition of p65-induced NF-κB promoter activity"

"TGFβ acts on the control of NF-κB: TNFα stimulation of TGFβ pretreated cells results in restricted NF-κB target gene expression caused by impaired p65 nuclear translocation"<-Thus we might have to upregulate NF-kappaB to get around the negative feedback that TGF-Beta exerts on NF-kappaB.

Inhibiting PKCDelta is a height increase target as well according to Inhibition of protein kinase Cdelta reduces tristetraprolin expression by destabilizing its mRNA in activated macrophages.

It's possible that you may want inflammation when performing LSJL to induce NF-kappaB decrease in let-7. This would involve longer durations of LSJL to induce more inflammation.


Overexpression of HMGA2-LPP fusion transcripts promotes expression of the alpha 2 type XI collagen gene.

"In a subset of human lipomas, a specific t(3;12) chromosome translocation gives rise to HMGA2-LPP fusion protein, containing the amino (N)-terminal DNA binding domains of HMGA2 fused to the carboxyl (C)-terminal LIM domains of LPP. HMGA2-LPP promotes chondrogenic differentiation, a marker of which is transactivation of the alpha 2 type XI collagen gene (Col11a2).  HMGA2-LPP and COL11A2 were co-expressed. either of HMGA2-LPP, wild-type HMGA2 or the N-terminal HMGA2 transactivated the Col11a2 promoter in HeLa cells, while the C-terminal LPP did not. HMGA2-LPP transcripts in lipomas with the fusion were 591-fold of full-length HMGA2 transcripts in lipomas without the fusion. in vivo overexpression of HMGA2-LPP promotes chondrogenesis by upregulating cartilage-specific collagen gene expression through the N-terminal DNA binding domains."

"only lipomas associated with HMGA2-LPP contained detectable COL11A2 transcripts"

"overexpression of the N-terminal DNA binding domains of HMGA2 in transgenic mice or constitutionally rearranged human results in overgrowth and lipomatosis"

"full-length HMGA2 can activate Col11a2 promoter when overexpressed. HMGA2 modulates the activity of the DNA repair gene ERCC1{down in LSJL} and of the cyclin A gene by binding to these genes"


"recombinant HMGA2 protein enhances the proliferation of porcine chondrocytes grown in vitro"

"statistically significant increase in proliferation could be detected in chondrocytes treated with either HMGA2-A peptides at both 10 μM (1.94x increase) and 50 μM (1.88x increase), or wild-type HMGA2 protein (10 μM, 2.1x increase){this was the only dose of HMGA2 used}." 




"LIN28B and its homolog LIN28A are functionally redundant RNA-binding proteins that block biogenesis of let-7 microRNAs. lin-28 and let-7 were discovered in Caenorhabditis elegans as heterochronic regulators of larval and vulval development but have recently been implicated in cancer, stem cell aging and pluripotency. The let-7 targets Myc, Kras{down in LSJL}, Igf2bp1 and Hmga2{up} are known regulators of mammalian body size and metabolism. we engineered transgenic mice to express Lin28a and observed in them increased body size, crown-rump length and delayed onset of puberty. These transgenic mice [had] increased glucose metabolism and insulin sensitivity."



"Lin28a Tg mice possess increased body size, a phenotype associated with genetic variation in the human LIN28B locus"



"Lin28a has been shown to enhance protein translation of Igf2, whose loss of imprinting causes a human overgrowth disorder called Beckwith-Wiedemann Syndrome (BWS). We found a ~20-fold increase of Igf2 mRNA in liver and a 2-fold increase in muscle. To determine if Igf2 was driving overgrowth, we crossed Lin28a Tg females to Igf2 null males and noted that Lin28a Tg mice lacking Igf2 were still overgrown "



"LIN28B alleles associated with later menarche were linked to taller stature"

Genetic variation in candidate genes like the HMGA2 gene in the extremely tall.

"The HMGA2 gene SNP [is] significantly associated with tall stature. Carrying the HMGA2 (rs1042725) C allele significantly increased the odds of being tall . In addition, controls with one or two copies of the C allele were significantly taller than controls carrying the TT genotype.
A common polymorphism in the HMGA2 gene is not only associated with height variation in the general population but also plays an important role in one of the extremes of the height distribution."

"The VDR gene encodes a nuclear receptor for 1,25-dihydroxyvitamin D. One study observed significant linkage for a functional SNP in its gene that may be responsible for 34% of idiopathic short stature cases"

"In humans, each copy of the C allele of this SNP was associated with an increase of about 0.4 cm in height in the general population"

Insights into the regulation of a common variant of HMGA2 associated with human height during embryonic development.

"genome-wide SNP studies revealed a significant association of rs1042725 genotypes CT and CC in the 3' UTR of HMGA2 with human height. HMGA2 expression levels during prenatal development might be a critical factor that contributes to the height phenotype. The rs1042725 genotype is unlikely to affect HMGA2 levels in pluripotent human embryonic stem cells (hESCs). hESCs in the inner cell mass of blastocysts are most likely not involved in determining the human height phenotype associated with this SNP. miR-196b as a candidate microRNA that could contribute to SNP-specific expression of HMGA2 during human prenatal development."

"HMGA2 is highly expressed in pluripotent human ES (hES) cells and contributes there to cell proliferation control. Variations in HMGA2 expression levels during these earliest stages of human development might influence adult stature. Based on the location of rs1042725 in the 3’UTR of HMGA2, microRNAs (miRs) play a role in HMGA2 regulation"

C-Terminal allele is the allele that's good for height.  You want CC.

"miR-196b [is] a candidate that specifically targets the C-allele of rs1042725 in a MRE and compromises expression of a reporter gene. miR-196b affects expression of Hoxb8 and Hoxa7 genes, which are involved in vertebrate limb formation during embryonic development"

"the presence of C-alleles at rs1042725 will lead to’reduced HMGA2 levels when compared with cells carrying T-alleles. Significantly increased body height is usually observed in mice and man when the HMGA2 gene is constitutively overexpressed during embryonic development and, later, in adult tissues of the body due to germline genomic alterations{so if LSJL can upregulate HMGA2 in the bone it could still help for adult height growth}. It is important to recognize that this is clearly an abnormal situation, which does not reflect normal development in a wild-type HMGA2 background when the temporal control of HMGA2 expression levels in, for example, specific progenitor stem cells might impact on body height.   Reduction of HMGA2 levels in combination with changes in expression patterns of other genes, such as certain Hox genes, might eventually lead to an increased body height. HMGA2 levels decrease during ESC differentiation, while miR-196 expression increases"

LIN28A Is a Suppressor of ER-Associated Translation in Embryonic Stem Cells.

"LIN28A binds to a large number of spliced mRNAs. LIN28A recognizes AAGNNG, AAGNG, and less frequently UGUG, which are located in the terminal loop of a small hairpin. LIN28A is localized to the periendoplasmic reticulum (ER) area and inhibits translation of mRNAs that are destined for the ER, reducing the synthesis of transmembrane proteins, ER or Golgi lumen proteins, and secretory proteins."

"Lin28a is highly expressed in embryonic stem cells (ESCs) and [is] one of the four factors that convert fibroblasts into induced pluripotent stem cells.  In [mice], Lin28a deficiency caused undergrowth in early stages of development, whereas its ectopic expression induced overgrowth and delayed the timing of puberty"

"LIN28 binds to the primary transcript of let-7 (pri-let-7) and prevents its processing by RNase III DROSHA. In the cytoplasm, it interacts with the precursor form of let-7 (pre-let-7) and interferes with pre-let-7 processing. LIN28 recruits TUTase 4 (ZCCHC11) to induce oligo-uridylation of pre-let-7, which effectively blocks DICER processing and facilitates degradation of the RNA. Although LIN28B is localized mainly in the nucleolus and interferes with nuclear processing, LIN28A is found mostly in the cytoplasmic compartment and acts in concert with TUTase 4. LIN28 homologs commonly have two types of RNA binding domains: a cold shock domain and a cluster of two CCHC-type zinc finger motifs. The “GGAG” sequences in the terminal loop of let-7 precursors serve as the binding site for the zinc finger domains that are critical for let-7 regulation"

"LIN28A can bind to and enhance translation of certain mRNAs such as Igf2 in differentiating myoblasts and Oct4 in ESCs "

"In mESCs, the let-7 family is likely to be the only functional miRNA target of LIN28A."

"LIN28A is a positive regulator of translation for mRNAs Igf2, cyclin A, cyclin B, histone 2a, and Oct4"

"The mRNAs coding LAMP1, EpCAM, and E-cadherin interact with LIN28A"

" LIN28A targets mRNAs for translational repression."  LIN28A represses all of the let7 family as verified by LIN28A knockdown studies.

12q14 microdeletion associated with HMGA2 gene disruption and growth restriction.

"The 12q14 microdeletion syndrome is a rare condition that [is] characterized by pre- and postnatal growth restriction, proportionate short stature, failure to thrive, developmental delay, and osteopoikilosis. Microdeletions within this region have ranged in size from 1.83 to 10.12 Mb with a proposed 2.61 Mb smallest region of overlap containing the LEMD3, HMGA2, and GRIP1 genes. [We identified] a 12q14 microdeletion in a female child presenting with proportionate short stature, failure to thrive, and speech delay. The genomic loss (minimum size 4.17 Mb, maximum size 4.21 Mb) contained 25 RefSeq genes including IRAK3, GRIP1, and the 3' portion of the HMGA2 gene.  partial deletion of HMGA2 [is] associated with the 12q14 microdeletion syndrome.  LEMD3 deletions [associates] with the 12q14 microdeletion syndrome. [HMGA2 has a role in human growth]."

Short stature is proportional leading weight to the theory that HMGA2 increases height by transcriptional repression of embryonic development leading to longer embryonic development and thus greater height growth.  All cases of this genomic deletion had proportional short stature which leads credence to the theory that HMGA2 affects height at the embryonic stage.

Probing into the Biological Processes Influenced by ESC Factor and Oncoprotein HMGA2 Using iPSCs.

"The high mobility group AT-hook 2 (HMGA2) protein is a nonhistone chromatin factor normally expressed in ESCs and during early developmental stages.  We used iPSC formation in conjunction with exogenous human HMGA2 expression. Anatomical development and cell adhesion/differentiation processes are strongly affected by HMGA2.  Expression of key diabetes susceptibility genes is influenced by HMGA2, which revealed an interesting link to the recently indentified Lin28/let-7 pathway regulating mammalian glucose metabolism. HMGA2 is not involved in the regulation of telomerase gene expression. Tight regulation of intracellular HMGA2 levels is important both to maintain a pluripotent ESC state and to induce differentiation into certain cell lineages during later developmental stages."

"In many tumor cells, including tumor initiating cells, HMGA2 is re-expressed as a result of Lin28-mediated let-7 miRNA degradation"

"HMGA2 exerts effects during pre- and postnatal development. miR-196b [is] involved in SNP-specific regulation of HMGA2 expression, during prenatal development, which is linked to an increased body stature in humans"

"Expression of HMGA2 in fibroblasts, which do not express HMGA2, promotes cell cycle arrest"

HMGA2 transgene iPSCs expressed less Utf1, Nr5a2, Oct4, Nanog, and Sox2 versus controls.

"HMGA2 expression [does not] influence iPSC morphology or affect some of the pluripotency phenotypes, although a slight decrease in iPSC colony number was detected."

"HMGA2 knockdown in human ESC showed a down-regulation of Nes"

"Igf2bp2, Irs1 and PiK3ip1 were up-regulated in cells with high HMGA2 levels"

"Prdm9, Dcdc2, Fcgbp, Fam107b and Clgn were up-regulated in H1 and H2 [HMGA2 transgene] cell lines"

"HMGA2 expression levels [are] critical both for maintenance of pluripotency and differentiation."

It should be noted that EFGP transgene was silenced after passage 8.

Genes upregulated by H1 and H2(these cell lines had the most significant increase in HMGA2 levels) and were differentially regulated by all three cell lines H1-H3 also upregulated by LSJL:
Dcdc2a{down}
Ccl7
Fos
Cxcl1
Egr1
Ccl2
Mmp3
Col3a1
Adamts1
Car12
Slurp1
Masp1
Junb
Col6a2{up and down}
Cthrc1
Col6a1
Apod
Barx2
Sod3
Lum
Gem
Cyr61
Fndc4
Fosb
Egr2
Odz3
Itgbl1
Thbs2
Smad9
Slitrk6
Eln
Mamdc2
Asph
Dkk3
Edn1
Bmpr1b
Mal2
Gas1
Sema3e
Htr2b
Aspn
Col5a2
Zfp36
Postn
1700001K23Rik
Maff
Lama4
Ecm2
Prrx1
Ssxb2
Col10a1
BMP2
BC051070{down}
Cacna2d2{down}
Ebf3{down}
D11Bwg0517e{down}
Rnf130{down}
Dcdc2a{down}
Usp29{down}
Cxcl5{down}
Fbxl22{down}
Xdh{down}
Usp29{down}
Ephx1{down}
Ninj1{down}
Sorbs2{down}
Rjl1{down}
Bcl11b{down}
C130026I21Rik{down}
Chi3l3{down}
Irs1{down}
Mafb{down}
Rgs10{down}
Gls{down}
P2ry14{down}
2010007H06Rik{down}
Cldn13{down}

Downregulated:
Steap2{up}
C77370{up}
Steap1{up}
Car8{up}
Brdt{up}
Stk32a{up}
Ccdc3{up}
Ttc26{up}
Car8{up}
Ttc26{up}
Scn3a{up}
Slco2a1{up}
Accn1
Cdx1
Mapk4
Sema7a
2410131K14Rik
Icam2
Gsta1
Rapgef3
5430407P10Rik
Arl11