Author Archives: admin


Hands across the water as bio bonds stand test of time | Business Weekly – Business Weekly

Boris Johnsons doomed reliance on Donald Trump to deliver a massive US trade deal for the UK is in stark contrast to the biotechnology bonds that continue to endure with massive commercial payback across the Atlantic.

Cambridge-US life science collaborations and investments have hit record levels in the last year and seemed destined to gather further pace through 2021 and beyond.

The Cluster now has GW Pharma and Bicycle Therapeutics under their own steam, Horizon Discovery (courtesy its acquisition by PerkinElmer), F-star (via its merger with Spring Bank) and Kymab (through its acquisition by Sanofi) quoted on Nasdaq, the US technology exchange. So lets delve deeper into the special relationship between UK and US biotechs that is not sullied by political fluctuations.

We reported in February 2020 that Bicycle Therapeutics clinched a $30 million upfront payment from Roche Group business Genentech as part of a strategic technology collaboration that could haul in up to $1.7 billion.

Bicycle will also be eligible to receive tiered royalties on Bicycle-based medicines commercialised by Genentech. Bicycle will be exploring its technology on a wider range of immuno-oncology targets, combining the expertise of both companies.

Bicycle will contribute its proprietary discovery platform, which allows rapid screening of novel targets to identify Bicycles and the ability to readily conjugate these together to create novel molecules that may overcome the potential limitations of other modalities. Genentech brings to the table its knowledge of immuno-oncology drug discovery and emerging target biology, as well as its development and commercialisation expertise.

In April 2020 and in another world first for a Cambridge life science business CellCentric revealed that it had developed CCS1477 the first p300/CBP inhibitor of its kind for use in the treatment of multiple cancer types.

Expanding on its UK-based clinical activities, the company disclosed that it was set to open US clinical sites for patient recruitment and had signed a milestone agreement with the Prostate Cancer Clinical Trial Consortium, LLC headquartered in New York to help select and manage US sites for the ongoing evaluation of CCS1477.

Also last April, Mogrify in the UK and Sangamo in the US agreed a collaboration and exclusive licence agreement for Mogrifys iPSC and ESC-derived regulatory T cells. It is a fistful of dollars play for startup business Mogrify which aims to transform the development of cell therapies by the systematic discovery of novel cell conversions; Nasdaq-quoted Sangamo Therapeutics is a genomic medicine company.

The deal allows Sangamo to develop allogeneic cell therapies from Mogrifys proprietary induced pluripotent stem cells and embryonic stem cells and Sangamos zinc finger protein gene-engineered chimeric antigen receptor regulatory T cell (CAR-Treg) technology.

In May, global food nutrition gamechanger Cargill headquartered in Minneapolis and Cambridge-based Eagle Genomics agreed a multi-year platform engagement to enable the digital transformation of microbiome and life sciences R & D across Cargills international territories. The deployment of the Eagle Genomics e[datascientist] platform will initially enable Cargills Health Technologies business to organise and synthesise additional insights from microbiome data amassed by the company over the past decade.

We announced in June that a young but big hitting US life science player was growing a UK team from a new base at Granta Park in Cambridge. Alloy Therapeutics, formed in Massachusetts in 2017, is dedicated to empowering global scientists with foundational drug discovery platforms and services. It is hiring fresh talent at its new UK home at Grantas McLintock Building.

The executive team is power-packed with business builders and money magnets. Led by founder, CEO and chairman Errik Anderson, the company is pioneering technology in a key growth area of the life sciences. Its specialist Discovery Services teams in Boston, MA and Cambridge, UK do immunisations every day while working to refine and improve the companys groundbreaking Alloy-Gx platform.

The sister Alloy Discovery Services business enables Alloy to ensure that all its partners are successful in their antibody discovery projects.

Also in June, Sosei Heptares set up a potential $409m exclusive discovery collaboration deal with New York-quoted AbbVie and there could be more cash cream to pour on top.

Sosei Heptares is eligible to receive up to $32m in upfront and near-term milestone payments as well as potential option, development and commercial milestones of up to $377m plus tiered royalties on global commercial sales.

Inivata, a Cambridge UK leader in liquid biopsy, reported in July that NeoGenomics, Inc its strategic commercialisation partner in the United States for the InVisionFirst-Lung liquid biopsy had commercially launched the test in the US.

InVisionFirst-Lung is a ctDNA next-generation sequencing liquid biopsy assay testing 37 genes relevant to the care of advanced non-small cell lung cancer. Nasdaq-quoted NeoGenomics, based in Florida, is a leading US-based cancer diagnostics and services company anchored in Florida.

July was also a seminal month for Acacia Pharma as the FDA approved its BYFAVO (remimazolam) for injection for the induction and maintenance of procedural sedation in adults undergoing surgical procedures lasting 30 minutes or less.

Acacia in-licensed the commercial rights to BYFAVO for the US from Cosmo Pharmaceuticals NV in January.

Avacta Group revealed in August that it could earn anything from $1/2bn to $1bn from an extended partnership with LG Chem Life Science, the Massachusetts-based subsidiary of the South Korean LG Group. The windfall stems from additional drug development programmes utilising Avactas Affimer XT technology.

Also in August, a US company personalising immune therapies for solid tumour cancers raised further capital to complete the Phase 1 buildout of a UK manufacturing facility in Sawston, Cambridge.

Northwest Biotherapeutics in Maryland raised $5 million growth capital and topped this up by securing a special purpose competitive loan of $1.77m from the Department for Business, Energy & Industrial Strategy. Northwest is developing DCVax personalised immune therapies for solid tumour cancers and is pursuing an intensive program of manufacturing preparations and planning as it approaches top line data from its Phase III trial.

In September, PhoreMost, the Cambridge biopharma company dedicated to drugging undruggable disease targets, and XtalPi Inc., a leading algorithm-driven AI-based pharmaceutical technology company, entered into a drug discovery collaboration agreement.

PhoreMost and Massachusetts-based XtalPi intend to rapidly identify and develop compounds to advance a drug discovery program against targets that epigenetically regulate tumour progression, and have been previously classified as undruggable.

A collaboration between Cambridge business Evonetix and Massachusetts company Analog Devices Inc was unveiled in September to fast-track the development and scale-up of the UK innovators desktop DNA writer. The technology will help facilitate the rapidly growing multibillion-dollar synthetic biology industry, Evonetix says.

Sosei Heptares earned more megabucks in September from its ongoing collaboration with $52 billion turnover New York-rooted pharma giant Pfizer. Pfizer reported that the first subject in a clinical trial had been dosed with a new drug candidate nominated from the multi-target drug discovery collaboration between the companies.

The milestone triggered a payment of $5 million to Sosei Heptares. The candidate was nominated for advancement by Pfizer in June 2019 generating a $3m milestone payment at that time.

Abzena pumped $60m into a new San Diego manufacturing hub in October a move that created 125 additional jobs with more to come.

Also last autumn, Cambridge biopharma business Arecor announced that it was partnering with global big-hitter Hikma Pharmaceuticals PLC to co-create a ready-to-administer injectable medicine in the US.

The breakthrough technology is being delivered through the quoted UK giants affiliate Hikma Pharmaceuticals USA Inc.

In December, Abcam opened a new 16,000 sq ft purpose-fitted facility in Fremont, CA, which will serve as a major new site for its specialist cell engineering team.

Also before Christmas, Owlstone Medical revealed that it was establishing a permanent office on the North Carolina Research Triangle Park and was hiring on both sides of the Atlantic.

Read more from the original source:
Hands across the water as bio bonds stand test of time | Business Weekly - Business Weekly

Exercise mimetics and JAK inhibition attenuate IFN-induced wasting in engineered human skeletal muscle – Science Advances

INTRODUCTION

Skeletal muscle comprises 45% of the total human body mass, and its contractile function is fundamental for the maintenance of life. Healthy skeletal muscle has a robust capacity to regenerate in response to minor injuries via activation, proliferation, and differentiation of muscle stem cells, a process that is greatly aided with the local and systemic inflammatory response (1). Interferon- (IFN-), in particular, is an important proinflammatory cytokine that regulates myogenic process during muscle regeneration, and its production is well balanced among natural killer (NK) cells, CD4+ and CD8+ T cells, and regulatory T cells (2). While inflammation following muscle injury promotes muscle regeneration, unregulated inflammatory reactions in many diseases, including chronic obstructive pulmonary disease, rheumatoid arthritis, dermatomyositis, cachexia, or sarcopenia, are associated with muscle loss and weakness (36). For example, sarcopenia is an age-associated syndrome characterized by progressive and generalized loss of skeletal muscle function and is usually associated with elevated expression of cytokines, including IFN- (6). In addition, elevated IFN- levels are routinely observed after influenza virus infection (7) and are associated with a cytokine storm resulting in tissue and organ damage in COVID-19 (8).

Besides numerous studies in rodents that show myopathic effects of inflammation in general and IFN- in particular (914), clinical studies have suggested that elevated IFN- in chronic inflammation and autoimmune diseases is one of major contributors to human skeletal muscle wasting and dysfunction (5, 6, 15). In a handful of in vitro studies in human muscle cells, IFN- has been shown to increase expression of human leukocyte antigenDR isotype (HLA-DR) class II antigens (16), proteases cathepsin B and L (17), and cytokines CXCL9, CXCL10, interleukin-6 (IL-6), and transforming growth factor (TGF-) (18, 19), as well as to inhibit myoblast growth and fusion (20). On the other hand, specific functional consequences and underlying mechanisms of IFN- elevation in human skeletal muscle have not been previously studied in vitro due to shortcomings of two-dimensional (2D) muscle cell culture (21). Recently, we have reported the first generation of 3D tissue-engineered models of functional human skeletal muscle (myobundles) made using primary myoblasts or induced pluripotent stem cellderived myogenic progenitors (2224). In optimized culture conditions, 3D human myobundles show superior myotube differentiation than 2D cultures and recapitulate the hallmark functional properties of native skeletal muscle including electrically or chemically induced twitch and tetanic contractions, a positive force-frequency relationship, robust calcium transients, and physiological responses to a diverse set of chemicals and drugs.

In more recent studies, we have shown that myobundles exhibit physiological response to exercise-mimetic electrical stimulation (E-stim) as evidenced from increased contractile strength, bulk muscle size, myotube diameter and length, sarcomeric protein expression, and glycolytic and fatty acid metabolism (24). In humans, E-stim and physical exercise can increase muscle mass and strength (25) and provide therapeutic benefits in the setting of chronic inflammation and aging (2628). The anti-inflammatory effects of muscle exercise are usually attributed to paracrine action of working myofibers on nonmuscle cells including adipocytes, macrophages, T cells, and NK cells (29). On the other hand, whether exercised myofibers can exert cell-autonomous anti-inflammatory effects on muscle structure and function has not been previously studied.

Here, we used the human myobundle system devoid of complex organ-organ interactions present in vivo to first time explore direct effects of IFN- on human skeletal muscle structure, function, and cytokine secretion and to unveil potential muscle-autonomous mechanisms underlying anti-inflammatory roles of exercise. We found that IFN- treatment induced muscle atrophy and reduced contractile function in human myobundles via up-regulation of the JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway (30), an effect that was directly countered by exercise-mimetic muscle stimulation. We further showed that the block of JAK/STAT up-regulation by IFN- with small-molecule JAK inhibitors tofacitinib (31) and baricitinib (32), clinically used for rheumatoid arthritis and in trials for COVID-19 (33), fully prevented muscle wasting and weakness induced by IFN-. Our findings establish human myobundles as a novel in vitro platform for studies of inflammatory muscle disease.

On the basis of previous studies in mice (9), we set to explore whether chronic 7-day application of a proinflammatory cytokine IFN- (20 ng/ml) to human myobundles cultured in serum-free media would lead to induction of muscle atrophy and weakness. Human myobundles made using cells derived from three independent donors were differentiated for 1 week, then exposed to IFN- for 1 week, and assessed for changes in cytokine secretion, structural, biochemical, and functional properties (Fig. 1A). To assess whether exercise-mimetic activity of myobundles can reduce expected myopathic effects of IFN-, we simultaneously applied chronic intermittent E-stim regime shown to increase myobundle mass and strength in our previous study (Fig. 1B) (24). In isometric force tests, we found that compared with the untreated control, IFN- treatment reduced myobundle twitch and tetanic force amplitude by 66 and 68%, respectively (Fig. 1, C, D, and G), while applying E-stim to untreated myobundles doubled their strength (Fig. 1, C, E, and G). E-stim cotreatment with IFN- significantly increased myobundle contractile force compared with IFN-only application (Fig. 1, D, F, and G), with twitch and tetanus amplitudes approximating those of untreated myobundles exposed to E-stim (Fig. 1, C, F, and G). These functional results were highly reproducible for myobundles from all three donors (fig. S1, A to C), as was the lack of any effects of IFN- or E-stim on passive tension measured at different stretch levels (fig. S1, D to F). Furthermore, measurements of force kinetics revealed that IFN- slowed both contraction (Fig. 1H) and relaxation (Fig. 1I) of myobundles and that E-stim rescued the IFN-induced force relaxation deficit as evidenced from the return of twitch recovery time (RT1/2) to control value (Fig. 1I). The exact numerical results of functional measurements are presented in table S1. Together, these studies showed that IFN- exerts adverse, inflammatory effects on contractile function of human myobundles and that exercise-mimetic activity can prevent IFN-induced muscle weakness.

(A) Schematic overview of myobundle culture, treatment, and characterization. Human primary myoblasts were expanded in 2D culture and mixed with hydrogel to form 3D myobundles, which were cultured in growth media (GM) for 4 days, then in differentiation media (DM) for 7 days, after which E-stim and/or IFN- (20 ng/ml) was applied for an additional 7 days. (B) E-stim protocol consisted of alternating 1-hour stimulation (S) at 10 Hz and 7-hour rest (R). (C to F) Representative twitch (Twi, 1 Hz) and tetanus (Tet, 20 Hz) force traces from myobundles (C) without IFN- or E-stim (IFN-, E-stim), (D) IFN-+, E-stim, (E) IFN-, E-stim+, and (F) IFN-+, E-stim+. (G) Twitch and tetanic force amplitudes averaged over three independent donors and shown relative to the IFN-, E-stim group (n = 5 to 10 myobundles per donor, 20 to 23 myobundles per group). (H and I) Time-to-peak tension (H; TPT) and half-relaxation time (I; 1/2RT) derived from contractile force recordings in myobundles (n = 47 to 75 data points from 20 to 23 myobundles from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

We next investigated mechanisms underlying IFN-induced myobundle weakness and functional benefit of E-stim, including possible changes in muscle structure, cytokine secretion, and calcium handling. From cross-sectional stainings (Fig. 2, A to C, and fig. S2, A to C), we found that both IFN- and E-stim similarly increased myobundle cross-sectional area (CSA) by ~40%, while E-stim of IFN-treated myobundles did not induce additional CSA increase (Fig. 2D). Similar to our previous study (24), the number of nuclei in myobundle cross section was significantly increased by E-stim in the absence of IFN- (Fig. 2E), while application of IFN-, with or without E-stim, resulted in unchanged nuclear numbers. Total F-actin+ area, a measure of myobundle muscle mass, was not altered by IFN-, indicating that IFN-induced force deficit was not a result of total muscle mass loss (Fig. 2F). On the other hand, E-stim induced muscle mass increase, albeit more in untreated (F-actin+ area, 0.56 mm2) than in IFN-treated (0.43 mm2) myobundles (Fig. 2F). We then assessed possible alterations in the myotube size by staining myobundle cross sections for dystrophin, a myotube membrane protein (Fig. 2C and fig. S2C). The IFN- treatment significantly reduced myotube diameter (11.3-m untreated versus 8.8-m IFN- treated) and area (fig. S2D), thus inducing myotube atrophy, whereas E-stim induced myotube hypertrophy by increasing myotube diameter and area in untreated and, to less extent, IFN-treated myobundles (Fig. 2G and fig. S2D). We further examined density of myotubes [labeled by F-actin and sarcomeric -actinin (SAA)] in myobundle cross sections and found that IFN- decreased myotube density, which remained at control levels in the presence of E-stim (fig. S2, E and F). Numerical results are presented in table S1. Together, these analyses suggest that IFN-induced myotube size and density decrease were prevented in exercise-mimetic E-stim conditions. We also assessed specific force of myobundles (contractile force normalized by muscle CSA), and similar to effects on myotube size and density, we found that E-stim prevented IFN-induced force decrease (Fig. 2H).

E-Stim prevents IFN-induced structural deterioration of myobundles. (A to C) Representative myobundle cross sections immunostained for (A) F-actin (green; scale bars, 500 m), (B) sarcomeric -actinin (SAA; red; scale bars, 500 m), and (C) dystrophin (Dys; red; scale bars, 25 m). (D to G) Quantification of (D) myobundle cross-sectional area (CSA), (E) number of nuclei per cross section, (F) F-actin+ area, and (G) myotube diameter [n = 19 to 27 images from three donors per group, 3 to 5 myobundles for each donor for (D) to (F), n > 900 myotubes from three donors per group for (G)]. (H) Force amplitude normalized per myobundle CSA (specific force, n = 20 to 23 myobundles from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+, **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

We also cryosectioned myobundles longitudinally and immunostained them for F-actin and SAA to assess potential changes in myotube and sarcomere organization across the entire myobundle depth (Fig. 3, A and B, and figs. S3 and S4). Adverse effects of IFN- treatment were most apparent in the interior of myobundles as evident from the decrease in projected myotube length (L; Fig. 3C and fig. S3, A and B), alignment (movie S1), and sarcomeric organization with significantly reduced percentage of cross-striated myotubes (from 45% in IFN- to 15% in IFN-+ group; Fig. 3D and fig. S3, C to F). As in our previous study (24), E-stim significantly increased myotube length (Fig. 3C) and percentage of cross-striated myofibers in untreated and, to a less extent, in IFN-treated myobundles (Fig. 3D), preventing the deteriorating effects of IFN- on myobundle structure. We then assessed changes in expression of contractile proteins (Fig. 3E) in myobundles and found that dystrophin and SAA expression remained unaltered with IFN- and E-stim treatments (Fig. 3, E to G), while changes in total myosin light chain (MYL) and myosin heavy chain (MYH) expression followed the same trend as the changes in measured specific force (Fig. 3, E, H, and I). Specifically, MYL and MYH protein expressions were significantly reduced by IFN- treatment and were increased by E-stim in both untreated and, to a less extent, IFN-treated myobundles. Regulation of MYL expression by skeletal muscle MYL kinase is associated with muscle differentiation (34) and the kinetics and amplitude of contractile force generation (35), while increase in MYH is associated with muscle differentiation, hypertrophy, and increased strength (24). Together, these studies showed that functional deficit due to IFN- treatment and benefit from E-stim could be at least in part explained by their opposite effects on contractile protein expression and organization.

(A and B) Representative longitudinal myobundle sections immunostained for (A) F-actin (green; scale bars, 100 m) and (B) SAA (red; scale bars, 50 m). (C and D) Quantification of (C) projected myotube length (n > 350 myotubes from three donors per group) and (D) SAA cross-striation frequency (n 9 images from each donor, n 37 images per group). (E) Representative Western blots from a single donor showing expression of the sarcomeric proteins dystrophin, myosin heavy chain (MYH; MF20), SAA, and myosin light chain (MYL; F-5), with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serving as a loading control. (F to I) Quantification of Western blots averaged for three donors with protein abundance normalized to GAPDH and shown relative to IFN- group. *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+, **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

To determine whether IFN-induced force deficit may be additionally caused by impaired calcium handling, we measured myobundle Ca2+ transients using Fluo-8 AM dye (Fig. 4, A to D, and movie S2). IFN- treatment reduced calcium transient amplitude to ~60% of that in the untreated myobundles (Fig. 4E and table S1). E-stim caused a significant increase in Ca2+ transient amplitude in both untreated and IFN-treated myobundles, consistent with changes in contractile force generation (Fig. 1G). To further obtain molecular insights in altered calcium handling in myobundles, we analyzed expression of RYR1 and CASQ1, proteins involved in sarcoplasmic reticulum Ca2+ release and buffering (Fig. 4F). Consistent with the observed decrease in Ca2+ transient amplitude, RYR1 and CASQ1 expressions were significantly reduced following IFN- treatment (Fig. 4, G and H). E-stim prevented the IFN-induced effects, significantly increasing the expression of RYR1 and CASQ1 to and beyond the control levels in untreated myobundles (Fig. 4, G and H). Collectively, these results revealed that IFN-induced weakness in myobundles was caused by deficits in both myotube structure and calcium handling.

(A to D) Representative peak Fluo-8 AM fluorescence intensity during E-stim (1 and 20 Hz) showing amplitudes of Ca2+ transients; scale bars, 500 m. (E) Quantified amplitudes (F/F) of electrically stimulated Ca2+ transients based on Fluo-8 AM recording (n = 8 myobundles from one donor per group). (F) Representative Western blots for RYR1, CASQ1, and GAPDH (loading control). (G and H) Quantified (G) RYR1 and (H) CASQ1 protein expressions normalized to that of GAPDH and shown relative to IFN- group (n = 6 samples from three donors per group). (I) Representative Western blots for pSTAT1, STAT1, and GAPDH (loading control). (J to L) Quantified (J) STAT1, (K) pSTAT1, and (L) pSTAT1/STAT1 protein expressions normalized to that of GAPDH and shown relative to the IFN-+ group (n = 6 samples from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

Regarding the known roles of IFN- in secretion and sensing of various cytokines, we quantified secretome of myobundles cultured in serum-free media in response to IFN- and/or E-stim using a multiplex beadbased assay (fig. S5). We found that application of IFN- increased myobundle secretion of several proinflammatory cytokines, namely, IL-7, IL-12p70, CX3CL1, IL-18, and monocyte chemoattractant protein-1 (MCP-1), and reduced myobundle secretion of IL-8 and leukemia inhibitory factor (LIF). E-stim altered myobundle secretome consistent with a report on human myotubes electrically stimulated short-term in serum-containing media (36). The cotreatment of myobundles with E-stim and IFN- partially or fully reversed the IFN-induced effects on IL-12p70, IL-18, IL-8, and LIF and had no effects on MCP-1 or CX3CL1 secretion. Application of E-stim increased the myobundle secretion of IL-6 to a similar level with and without IFN- application, consistent with previous findings in mice suggesting that IFN-induced muscle wasting is IL-6 independent (9).

Previous studies have suggested that proinflammatory effects of IFN- in muscle result from up-regulation of the JAK/STAT pathway (37). We thus measured the protein expression of total STAT1 and phosphorylated STAT1 (pSTAT1) and assessed the degree of STAT1 activation defined as pSTAT1/STAT1 (Fig. 4, I to L). We found that IFN- treatment increased both the total STAT1 and pSTAT1 levels as well as the ratio of pSTAT1/STAT1. While E-stim had no significant effects on STAT1 or pSTAT1 levels in the absence of IFN-, it significantly attenuated IFN-induced pSTAT1 increase without affecting the levels of STAT1 (Fig. 4, I to L). Together, E-stim partially (~50%) attenuated the IFN-induced up-regulation of the JAK/STAT1 pathway in myobundles, which, in addition to direct effects of E-stim on muscle hypertrophy and strengthening, represents a novel, independent mechanism for the beneficial anti-inflammatory effects of exercise on muscle weakness and wasting induced by IFN-.

Beneficial effects of E-stim in conjunction with its partial down-regulation of JAK/STAT1 signaling prompted us to test in an independent set of experiments whether the direct inhibition of JAK/STAT pathway by Food and Drug Administration (FDA)approved small-molecule inhibitors tofacitinib (Tofa, blocker of JAK1/2/3) and baricitinib (Bari, blocker of JAK1/2) can prevent structural and functional deficits induced by IFN- (Fig. 5A). We found that 8-day treatment with either inhibitor (at a clinically relevant dose of 500 nM) had no adverse effects on myobundle morphology and function. On the other hand, the same dose of the inhibitors fully prevented the deteriorating effects of IFN- on myobundle contractile force and kinetics in all of the donors (Fig. 5B and fig. S6, A to F), as well as blocked the IFN-induced increase in myobundle CSA (Fig. 5C) and decrease in myotube size, length, and abundance of cross-striations (Fig. 5, D to F, and fig. S7, A to C). The exact numerical results of functional measurements are presented in table S2. We next investigated whether JAK/STAT inhibitors prevented the IFN-induced changes in myobundle contractile protein expression and calcium handling. Treatment of nave myobundles with the inhibitors had no apparent effects on contractile or calcium handling protein expression or generation of Ca2+ transients (Fig. 6, A to F, and fig. S7, D and E). Coapplication of JAK/STAT inhibitors with IFN- fully prevented the IFN-induced decrease in expression of MYL and MYH (Fig. 6, A to C), Ca2+ transient amplitude (Fig. 6D), and RYR1 and CASQ1 expression (Fig. 6, E and F), without any changes in dystrophin or SAA levels (fig. S7, D and E).

(A) Schematic overview of myobundle culture, treatment, and characterization. Human primary myoblasts were expanded in 2D culture and mixed with hydrogel to form 3D myobundles, which were cultured in GM for 4 days, then in DM for 6 days, after which JAK inhibitors (JAKi) tofacitinib (Tofa, 500 nM) or baricitinib (Bari, 500 nM) was applied for additional 8 days, the last 7 of which in the presence or absence of IFN-. (B) Contractile force amplitude per myobundle CSA (specific force, n = 11 to 15 myobundles from three donors per group). (C) Quantified CSA of myobundles. (D) Quantified myotube diameter from myobundle cross sections (n 720 myotubes from three donors per group). (E and F) Quantified (E) projected myotube length (n > 150 myotubes from two donors per group) and (F) SAA cross-striation frequency (n 30 images from three donors per group) from longitudinal sections of myobundles. *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

(A) Representative Western blots from a single donor showing expression of dystrophin, MYH (MF20), SAA, and MYL (F-5), with GAPDH serving as a protein loading control. Tofa + I: tofacitinib + IFN-, Bari + I: baricitinib + IFN-. (B and C) Quantifications of Western blots for (B) MYL and (C) MYH averaged for three donors with protein abundance normalized to GAPDH and shown relative to IFN- group (n = 6 samples from three donors per group). No difference in dystrophin or SAA expression was observed. (D) Quantified amplitudes of electrically stimulated (1 and 20 Hz) Ca2+ transients (n = 8 myobundles from one donor per group). (E) Representative Western blots for RYR1, CASQ1, and GAPDH. (F) Quantified RYR1 and CASQ1 protein expression normalized to that of GAPDH and shown relative to the IFN- group (n = 6 samples from three donors per group). (G) Representative Western blots for pSTAT1, STAT1, and GAPDH. (H and I) Quantified (H) pSTAT1 and (I) pSTAT1/STAT1 protein expression normalized to that of GAPDH and shown relative to the IFN-+ group (n = 3 samples from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+; **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

Because JAK/STAT inhibitors completely prevented the inflammatory action of IFN- on myobundles, we assessed their effects on STAT1 expression and activity by Western blotting. The treatment of nave myobundles with tofacitinib did not affect the expression of STAT1 or pSTAT1, while baricitinib moderately increased the expression of STAT1 but not pSTAT1 (Fig. 6, G to I, and fig. S7F), consistent with previous studies (38). Application of the inhibitors to IFN-treated myobundles significantly decreased pSTAT1 expression to near-nave levels (Fig. 6, G and H), without altering the IFN-induced increase in STAT1 expression (fig. S7F). Consequently, the IFN-induced increase in STAT1 activity (pSTAT1/STAT1) was fully prevented by coapplication of the inhibitors, explaining their strong protective effects on myobundle function. Together, these results established that the adverse effect of IFN- on human myobundles was predominantly mediated via up-regulation of JAK/STAT1 signaling rather than changes in alternative signaling pathways (39).

Here, we used a human myobundle system to model inflammation-induced muscle weakness and explore anti-inflammatory mechanisms of muscle exercise. Previously, we have shown that myobundles represent a unique personalized in vitro platform to study not only structural and biochemical but also metabolic and contractile responses of human skeletal muscle to a diverse set of pathological and physiologic inputs, including exercise (2224, 40). In this study, we sought to explore how human skeletal muscle strength and structure are affected by IFN-, a prototypical cytokine elevated in various inflammatory diseases (18, 41, 42). Consistent with studies in mice (13, 14), 7-day treatment of human myobundles with IFN- (20 ng/ml) induced muscle weakness (i.e., weaker and slower contractions) that was associated with significant myofiber atrophy and disarray, reduced expression of contractile and calcium handling proteins, and altered cytokine expression. These results were fully reproduced in myobundles derived from three independent donors. When coapplied with IFN-, a 7-day intermittent exercise-mimetic E-stim had pronounced protective effects on myobundles. Beside its well-established hypertrophic and strengthening effects (24, 43), the exercise-mimetic E-stim (24) partially reduced IFN-induced STAT1 up-regulation, established using selective JAK/STAT inhibitors to be the dominant proinflammatory mediator of IFN- action in myobundles. To our knowledge, this is the first study to explore direct and specific effects of IFN- on human skeletal muscle function and to demonstrate the existence of a novel myofiber-autonomous, anti-inflammatory mechanism of muscle exercise involving the JAK/STAT1 pathway (fig. S8). We anticipate the future use of myobundle platform to study human inflammatory disease and anti-inflammatory therapies.

Skeletal muscle is an important secretory organ in the body that both responds to systemic endocrine signals and can function as an active regulator of immune and inflammatory response. Physical exercise can alter the cytokine secretion of skeletal muscle and other tissues, which in turn can promote muscle repair after injury (44), inhibit muscle atrophy (26), and potentially act as an anti-inflammatory therapy in chronic inflammatory diseases (27). While the anti-inflammatory effect of exercising muscle fibers has been mainly attributed to their paracrine cross-talk with proinflammatory nonmuscle cells (29), the use of myobundles devoid of immune or fat cells in this study allowed us to unveil muscle-autonomous inhibitory effects of exercise on JAK/STAT1 signaling, a mediator of muscle inflammation caused by IFN-. This exercise-induced down-regulation of pSTAT1 could be autocrine mediated via the partial normalization of IFN-induced changes in myobundle secretome; however, pleotropic effects of muscle exercise on several intracellular signaling pathways (45) and their potential cross-talk with JAK/STAT signaling (39, 46, 47) warrant future investigations. Our studies also confirmed that the FDA-approved small-molecule JAK/STAT inhibitors tofacitinib and baricitinib, prescribed for rheumatoid arthritis, fully prevented IFN-induced muscle wasting at a nontoxic, therapeutically relevant dose (32), although their biochemical mechanisms of action may vary based on the somewhat differing effects on STAT1 and pSTAT1 expression.

In summary, we have applied an in vitro 3D myobundle model to investigate the interplay between IFN- and exercise-mimetic stimulation in inflammatory response of human skeletal muscle. We show that chronic application of IFN- induces myobundle weakness via up-regulation of JAK/STAT1 signaling pathway, which can be partially prevented by exercise-mimetic stimulation and fully prevented by treatment with clinically approved JAK/STAT inhibitors. We envision that incorporation of additional human nonmuscle cells in the myobundle platform [akin to use of macrophages in a rat skeletal muscle model (40)] multiplexed with application of various inflammatory cytokines (18), exercise-mimetic regimes (24), and candidate therapeutics will allow comprehensive mechanistic studies of human muscle inflammation, anti-inflammatory roles of exercise, and discovery of effective therapies for muscle wasting.

Human skeletal muscle samples were obtained from three donors (two females and one male, age 12 to 18) through standard needle biopsy or surgical waste from three donors with informed consent under Duke University Institutional Review Boardapproved protocols (Pro00048509 and Pro00012628). Muscle tissue was minced using sharp scissors and enzymatically digested with 0.05% trypsin for 30 min at 37C. Isolated cells were collected by centrifugation and resuspended in growth media [GM; low-glucose Dulbeccos modified Eagles medium (DMEM) (Thermo Fisher Scientific, D6046) supplemented with 10% fetal bovine serum (FBS) (Hyclone, SH30071.03), fetuin (50 g/ml) from FBS (Sigma-Aldrich, F2379), recombinant human epidermal growth factor (EGF) (PeproTech, AF-100-15), dexamethasone (Sigma-Aldrich, F2379), and penicillin G (100 U/ml, Thermo Fisher Scientific)] and preplated for 2 hours to reduce the number of fibroblasts. After preplating, the cells were transferred onto to 1% Matrigel (BD Biosciences)coated flasks, cultured in GM, and expanded by passaging after reaching 70% confluence. At passage 4 or 5, cells were detached, counted, and used to fabricate myobundles.

Human myobundles were fabricated as described previously (23, 24). Briefly, polydimethylsiloxane (PDMS) molds made to fit in a well of a 12-well plate were casted from 3D-machined Teflon masters containing two semicylindrical wells (7 mm long, 2-mm diameter for culture of two myobundles), sterilized in 70% ethanol, air dried in a tissue culture hood, and coated with 0.2% (w/v) Pluronic F-127 (Invitrogen) for at least 1 hour at room temperature to prevent tissue adhesion. Laser-cut Cerex frames (9 9 mm2, 1-mm-wide rim) were sterilized and placed within the air-dried PDMS molds. The frame provided an anchor surface for myobundle attachment and functioned as a mechanical guide during cell-mediated hydrogel compaction, resulting in uniaxial cell alignment (48). Expanded myogenic cells were dissociated using 0.025% trypsin-EDTA and encapsulated in a hydrogel solution at 1.5 107 cells/ml. For a single myobundle, the cell/hydrogel solution contained 10 l of bovine fibrinogen (20 mg/ml) in phosphate-buffered saline (PBS) (Sigma-Aldrich), 10 l of Matrigel (Corning), 2 l of bovine thrombin (50 U/ml) in 0.1% bovine serum albumin (BSA) in PBS (Sigma-Aldrich), and 28.2 l of GM with 0.75 106 cells, prepared on ice, which was mixed thoroughly and immediately pipetted into one well of the PDMS mold. Cell/hydrogel mixture in PDMS molds was polymerized at 37C for 30 min, and thereafter, myobundles were cultured on a rocker. After 2 days, frames with myobundles were removed from the molds and left to float in culture media. GM was used for the first 4 days of culture, after which it was replaced by differentiation medium [DM, low glucose DMEM (Thermo Fisher Scientific) supplemented with N2 supplement (100, Thermo Fisher Scientific), penicillin G (100 U/ml, Thermo Fisher Scientific)], with media changes performed daily. Media were supplemented with 6-aminocaproic acid (ACA; Sigma-Aldrich) to reduce fibrinolysis (1.5 mg/ml in GM. 2 mg/ml in DM).

For inflammation experiments, starting on differentiation day 7 (culture day 11), myobundles were treated for an additional 7 days with IFN- (20 ng/ml) [PeproTech, 300-02, in BSA final (0.5 g/ml)] freshly added during each media change. Control group was supplemented with media only. For E-stim experiments, we used custom-made PDMS chambers containing two parallel carbon electrodes, as previously described (24, 49). Myobundles were placed in the chamber, and E-stim was applied from differentiation days 7 to 14 either without or with IFN- treatment using a D330 MultiStim system (Digitimer Ltd.) controlled by a custom-made LabVIEW program. The applied intermittent regime of stimulation consisted of 1-hour stimulation cycles (bipolar 2 ms, 70-mA constant-current impulses applied in 0.5-s-long 10-Hz pulse trains delivered every 5 s) separated by 7-hour rests (Fig. 1B) (24). For experiments with JAK/STAT inhibitors, tofacitinib (in water; Sigma-Aldrich, PZ0017) or baricitinib (in 0.01% DMSO final; Sigma-Aldrich, G-5743,) was added at 500 nM concentration to myobundles starting on differentiation day 6, 1 day before IFN- treatment, and continued to be freshly applied (with or without IFN-) for the following 8 days during the daily media changes.

Contractile force generation and passive tension in myobundles were measured using a previously described custom-made force measurement apparatus (22). Briefly, single myobundles were transferred to the force measurement apparatus and immersed in DMEM media at 37C. One end of the myobundle was attached to a fixed PDMS block, and the other to a movable PDMS float connected to a force transducer mounted on a computer-controlled motorized linear actuator (Thorlabs, Newton, NJ). The sides of the frame were cut to allow force measurements. Using the linear actuator, the myobundle was stretched to 100, 108, and 116% of their original culture length, and twitch contractions were recorded in response to 10-ms electrical pulses applied by platinum electrodes at 1-Hz stimulation rate. At 16% stretch, 1-s-long stimulation at 20 Hz was applied to record tetanic contractile force. Force traces were analyzed for peak twitch and tetanus force, passive tension, time-to-peak twitch, and half-relaxation time using a custom MATLAB program (22, 40).

Myobundles were fixed in 2% (v/v) paraformaldehyde (Electron Microscopy Sciences) in PBS overnight at 4C with rocking. Fixed samples were submerged in optimal cutting temperature compound (Electron Microscopy Sciences) and snap frozen in liquid nitrogen. Constructs were sectioned (10 m thick) parallel (longitudinal) and perpendicular (cross) to the long axis of the bundles using a cryostat (LEICA CM1950) and were mounted onto glass slides. Before staining, sections were incubated in a blocking solution containing 5% chick serum and 0.1% Triton X-100 in PBS overnight at 4C. Primary antibodies were applied in blocking solution overnight at 4C. Secondary antibodies were applied overnight at 4C. Antibody information and dilutions are listed in table S3. Immunostained samples were mounted with ProLong Glass Antifade reagent (Thermo Fisher Scientific, P36984). Fluorescence images were acquired using an Andor Dragonfly spinning disk confocal microscope at 10 to 40 magnification and analyzed by ImageJ.

Images of myobundle longitudinal sections stained for F-actin and SAA were used for the measurement of myotube organization. Projected length of clearly distinguishable F-actinlabeled myotubes was determined manually (fig. S3, A and B). Percentage of cross-striated myotubes was assessed by manually counting the cross-striated myotubes labeled by SAA staining divided by the total number of myotubes determined from F-actin costaining of the same section.

Media conditioned by myobundles from three donors were collected between days 3 and 4 of E-stim for the E-stim groups and on equivalent culture days (between days 14 and 15) for the groups without E-stim, then frozen, and used for the secretome analysis on the same plate upon thawing. Select cytokine concentrations were measured using a custom-designed human magnetic 18-plex panel for the Luminex platform (Thermo Fisher Scientific, Waltham, MA) by the Immunology Core at Duke University following the manufacturers instructions.

Protein was isolated from three to four myobundles per experimental sample in ice-cold radioimmunoprecipitation assay lysis and extraction buffer in the presence of protease inhibitor (Sigma-Aldrich) and phosphatase inhibitor cocktail (Roche), as previously described (24). Protein concentration was determined using a bicinchoninic acid assay (Thermo Fisher Scientific). Western blots were performed using Bio-Rad Mini-PROTEAN 4 to 15% gradient gels with proteins transferred using a Bio-Rad Mini Trans-Blot Cell. After blocking with 5% milk or 5% BSA in tris-buffered saline with 0.1% Tween 20 at room temperature for 1 hour, primary antibodies diluted in the same blocking solution were incubated with the membrane at 4C overnight. Horseradish peroxidaseconjugated anti-mouse and anti-rabbit secondary antibodies were applied for 1 hour at room temperature. Antibody information and dilutions are listed in table S3. Protein detection was performed using either SuperSignal West Pico PLUS or Femto Maximum ECL chemiluminescence substrates (Thermo Fisher Scientific). Images were acquired using a Bio-Rad ChemiDoc imaging system and analyzed with ImageJ.

For Ca2+ transient measurements, myobundles were incubated with 50 M of calcium-sensitive dye Fluo-8 AM (Abcam, ab142773) in DM in an incubator for 1 hour while rocking, followed by washing in dye-free media for 30 min. Electrically induced Ca2+ transients were recorded as previously described (24, 40). Myobundles were transferred into a glass-bottom live-imaging chamber with Tyrodes solution warmed at 37C in a heated live-imaging chamber. Fluorescence images were acquired at 4 magnification on a Nikon microscope using a high speed EMCCD (electron multiplying charge-coupled device) camera (Andor iXon 860) and Andor Solis software. Ca2+ transient amplitudes were calculated as the maximum relative change in fluorescence signal, F/F = (Peak Trough)/(Trough Background).

Experimental data are presented as means SEM. Statistical significances between groups were verified by one-way analysis of variance (ANOVA) with Tukeys post hoc test or as described in the figure captions, using GraphPad Prism (GraphPad Software). P < 0.05 was considered statistically significant. Sample sizes for in vitro experiments were determined based on variance of previously reported measurements. All immunostaining images and movies shown are representative of similar results from at least three independent experiments.

Acknowledgments: We thank A. Khodabukus for technical advice and K. Huang for advice on writing the manuscript. Funding: Research reported in this study was supported by the NIH under grants UH3TR002142, U01EB028901, and R01AR070543. The content of the manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Author contributions: N.B., L.R., and Z.C. conceived the idea and designed the experiments; Z.C., L.R., B.L., and R.-Z.Z. performed the experiments and analyzed the data; Z.C. and N.B. wrote the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

See the original post:
Exercise mimetics and JAK inhibition attenuate IFN-induced wasting in engineered human skeletal muscle - Science Advances

Can Science Save the Northern White Rhino? – Freethink

The northern white rhino reproduces like most mammals: a male and a female have sex, the female gets pregnant, and about 18 months later, a new rhino calf is born.

But right now, there aren't any male northern white rhinos the last one, Sudan, died in 2018 and just two females remain.

In the past, that would mean the species' total extinction was just on the horizon.

Today, though, teams of researchers are exploring three different plans to revive the northern white rhino and one involves a controversial technique that could also revolutionize human reproduction.

The first plan for saving the northern white rhino is straightforward, science-wise: in vitro fertilization (IVF).

Prior to his death, Sudan lived at Ol Pejeta Conservancy, a protected wildlife area in Kenya, along with the only remaining females of his species, Najin and her daughter Fatu.

When Sudan and four other males were still alive, Ol Pejeta researchers collected samples of their sperm. They've since used some of that sperm to fertilize eggs they retrieved from Najin and Fatu in 2019.

They hope to inseminate the potential mothers before 2022.

This has resulted in five viable embryos.

Both Fatu and Najin have known reproductive issues that would prevent them from being able to carry a calf to term, so researchers plan to use female southern white rhinos a close relative of the northern white rhino, with numbers in the ten thousands as surrogates.

Their hope is to complete the insemination of the potential mothers before 2022.

However, no one knows for sure whether the pregnancy will take, as rhino reproduction is complicated. The first southern white rhino conceived through artificial insemination was only just born in July 2019, and no one has been able to produce one through IVF yet.

Southern white rhinos are also at the center of another plan to save their close relatives from extinction.

The procedure researchers developed to extract eggs from Fatu and Najin required the use of a risky anesthetic, so before subjecting the pair to it, they tested the technique on 12 southern white rhinos.

They then used sperm from northern white rhinos to fertilize some of the eggs they collected.

If any calves were born from those hybrid embryos, they'd be half northern white rhino and half southern, which would keep the former species somewhat alive.

Still, Fatu and Najin would be the last fully northern white rhinos, and their deaths would bring about the species' permanent extinction unless scientists go to Plan C, a very experimental (and sometimes controversial) technique called in vitro gametogenesis (IVG).

In addition to collecting sperm and eggs from the last northern white rhinos, researchers have also collected and frozen tissue samples from about a dozen members of the species.

In 2011, a group at Scripps Research Institute in California proved it was possible to create induced pluripotent stem cells (iPSC) from this rhino tissue. These cells can be prompted to grow into any type of specialized cell under the right conditions including reproductive cells.

In theory, researchers could prompt northern white rhino cells to develop into sperm and eggs, fertilize the eggs with the sperm, and then implant the embryos into surrogates (like the southern white rhinos).

Researchers have actually used IVG like this to successfully impregnate mice.

If it works in rhinos or other animals, it could bring whole species back from extinction (assuming you had enough tissue samples and a surrogate mother species).

If scientists can make it work in humans, it would completely change the parameters for reproduction. Single people could have babies related only to them, creating their own eggs and sperm, and same-sex couples could have tots that are related to them both (though two men would need to find a surrogate to carry the baby).

Again, IVG has only been shown in mice so far, so the use of such an experimental technique in rhinos would likely be years down the line (and humans even further).

However, it does offer hope that the northern white rhino species doesn't have to die with Fatu and Najin, even after their last egg is gone.

We'd love to hear from you! If you have a comment about this article or if you have a tip for a future Freethink story, please email us at [emailprotected].

Link:
Can Science Save the Northern White Rhino? - Freethink

Global Phenylketonuria (PKU) Treatment Market Detailed Analysis of Current Industry Figures with Forecast Growth By Top Competitors:American Gene…

A consistent statistical surveying report like this Phenylketonuria (PKU) Treatment report stretches out your reach to the achievement in your business. All the information and measurement remembered for the report is supported up by notable investigation devices which incorporate SWOT examination and Porters Five Forces investigation. Statistical surveying contemplates did in this report are chivalrous which help organizations to take better choices and create predominant methodologies about creation, advertising, deals and advancement. Market definition, market division, key improvements in the market, serious investigation and examination approach are the significant section of this Phenylketonuria (PKU) Treatment market report which are again explained accurately and explicitly.

PHENYLKETONURIA (PKU) TREATMENT MARKETis rising at substantial CAGR in the forecast period of 2019-2026. This rise in market can be attributed to increasing incidence rate of phenylketonuria, growing research in the field of genomics and biotechnology, increasing awareness about the phenylketonuria, change in lifestyle of the people, rising per capita income in emerging markets and development in the health care industry.

Ask For Complimentary Sample PDF| Request Athttps://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-phenylketonuria-pku-treatment-market

Few of the major competitors currently working in the global phenylketonuria (PKU) treatment market are

DAIICHI SANKYO COMPANY, LIMITED,Ajinomoto Cambrooke, Inc.,American Gene Technologies, Inc.,Ultragenyx Pharmaceutical,Danone Nutricia ,Reckitt Benckiser Group plc,Abbott,Promin Metabolics,Ajinomoto Cambrooke, Inc,Promin,Som innovation biotech,Synthetic Biologics, Inc..

Segmentation: Global Phenylketonuria (PKU) Treatment Market

By Type

(Hyperphenylalaninemia, Mild PKU, Moderate or Variant and Classic PKU),

Drugs Type

(Kuvan, Sapropterin, Palynziq, Pagvaliase, Biopten),

Therapy Type

(Gene Therapy, Dietary Therapy),

Route of Administration

(Oral, Intravenous, Subcutaneous and Others),

End- Users

(Hospitals, Homecare, Specialty Clinics, Others),

Geography

(North America, South America, Europe, Asia-Pacific, Middle East and Africa)

Enquire Here Get customization & check discount for report @:https://www.databridgemarketresearch.com/inquire-before-buying/?dbmr=global-phenylketonuria-pku-treatment-market

Market Drivers

Market Restraints

Key Developments in the Market:

Important Points Covered in Phenylketonuria (PKU) Treatment Market Report Are:

Access Complete Report @https://www.databridgemarketresearch.com/reports/global-phenylketonuria-pku-treatment-market

Key points mentioned

AboutData Bridge Market Research

An absolute way to forecast what future holds is to comprehend the trend today!

Data Bridge set forth itself as an unconventional and neoteric Market research and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best market opportunities and foster efficient information for your business to thrive in the market. Data Bridge endeavors to provide appropriate solutions to the complex business challenges and initiates an effortless decision-making process.

Data bridge is an aftermath of sheer wisdom and experience which was formulated and framed in the year 2015 in Pune. We ponder into the heterogeneous markets in accord with our clients needs and scoop out the best possible solutions and detailed information about the market trends. Data Bridge delve into the markets across Asia, North America, South America, Africa to name few.

Data Bridge adepts in creating satisfied clients who reckon upon our services and rely on our hard work with certitude. We are content with our glorious 99.9 % client satisfying rate.

Contact: Data Bridge Market Research Tel: +1-888-387-2818 Email:Corporatesales@databridgemarketresearch.com

Go here to read the rest:
Global Phenylketonuria (PKU) Treatment Market Detailed Analysis of Current Industry Figures with Forecast Growth By Top Competitors:American Gene...

Global MRD (Minimal Residual Disease) Testing Market Report 2020-2025: One of the Most Rapidly Evolving and Dynamic Markets – GlobeNewswire

January 22, 2021 07:18 ET | Source: Research and Markets

Dublin, Jan. 22, 2021 (GLOBE NEWSWIRE) -- The "Global MRD Testing Market: Focus on Technology, Application, End User, Region and Competitive Landscape - Analysis and Forecast, 2020-2025" report has been added to ResearchAndMarkets.com's offering.

The global market for MRD testing is predicted to grow at a CAGR of 15.64% over the forecast period, 2020-2025.

MRD Testing industry to be one of the most rapidly evolving and dynamic markets. The market is driven by certain factors, such as the rising incidence of hematologic malignancies, the increasing consumer awareness for tailored therapy, the increasing research funding from the National Cancer Institute, and the increasing disposable income in emerging economies.

The market is favored by the developments in the field of MRD testing solutions for hematologic malignancies and solid tumors. Currently, the MRD testing industry is witnessing an upsurge due to the rising incidence of hematologic malignancies, resulting in the high demand for sensitive testing solutions. Additionally, the high adoption of MRD tests among patients and the growing awareness among physicians regarding MRD testing are some of the critical factors expected to bolster the market growth.

Furthermore, diagnostic companies are focusing on the development of NGS-based MRD tests for lymphoid malignancies, having higher sensitivity and low turnaround time to benefit the patients suffering from hematologic malignancies.

Competitive Landscape

The exponential rise in the number of cases associated with hematological malignancies has created a buzz among the diagnostic companies to further invest in the development of reliable, sensitive, and rapid MRD testing solutions to aid patients to get into remission. Due to the presence of a diverse product portfolio and intense market penetration, Invivoscribe, Inc. has been a pioneer in this field and has been a significant competitor in this market.

On the basis of region, North America holds the largest share of the MRD testing market due to high infusion of funding from government organizations for conducting research on hematological malignancies, rising incidence of hematological malignancies, and high adoption of technologically advanced MRD tests, among others. Apart from this, Asia-Pacific is anticipated to grow at the fastest CAGR during the forecast period, 2020-2025.

Key Questions Answered in this Report:

Key Topics Covered:

1 Product Definition and Market Scope

2 Research Methodology

3 Market Overview 3.1 MRD Testing 3.2 MRD Testing: Solid Tumor vs Hematological Malignancies 3.3 Market Footprint 3.4 Market Size and Future Growth Potential

4 Market Dynamics 4.1 Impact Analysis 4.2 Market Drivers 4.2.1 Rising Incidence of Hematologic Malignancies 4.2.2 Increasing Consumer Awareness for Tailored Therapy 4.2.3 Increase in Research Funding from National Cancer Institute 4.2.4 Increasing Disposable Income in Emerging Economies 4.3 Market Restraints 4.3.1 False Negatives and Positives 4.3.2 Uncertain Reimbursement and Regulatory Policies 4.3.3 Lack of Trained Professionals 4.3.4 Lack of Established Treatment Protocols for High-Value Tests in Emerging Economies 4.4 Market Opportunities 4.4.1 Potential Long-Term Cost Savings 4.4.2 Increasing Market Access in Emerging Economies 4.4.3 Technological Evolution of Testing 4.4.3.1 8- and 10-Color Flow Cytometry 4.4.3.2 PCR for Gene Rearrangements 4.4.3.3 NGS and Multiplexing

5 Industry Insights 5.1 Approval Scenario 5.1.1 Approved Minimal Residual Disease Tests by Major Players 5.1.2 Launched Minimal Residual Disease Tests by Major Players 5.2 Financing Scenario 5.2.1 Key Players Stratification (as Per Raised Financing Value) 5.2.2 Key Players Financing Analysis (FY2017-2019) 5.3 Regulatory Framework 5.4 Reimbursement Scenario 5.5 Supply Chain Analysis 5.5.1 For Laboratory Developed Tests (LDTs) 5.5.2 For In-Vitro Diagnostics (IVDs) 5.6 Minimal Residual Disease Testing Government Initiatives 5.6.1 Technology Consideration for Minimal Residual Disease 5.6.1.1 Considerations for Cellular Technology Platforms 5.6.1.2 Considerations for Molecular Technology Platforms 5.6.1.3 Considerations for Sample 5.7 Impact of COVID-19 on Minimal Residual Disease Testing Market 5.7.1 Considerations With Inpatients 5.7.2 Considerations With Outpatients 5.8 Price Sensitivity Analysis (Elasticity) 5.8.1 Physicians' Perception 5.8.2 Investors' Perception 5.8.3 Payors' Perception

6 Global Minimal Residual Disease Testing Market: Competitive Insights 6.1 Overview 6.2 Synergistic Activities 6.3 Approvals 6.4 Product Launches and Updates 6.5 Other Developments 6.6 Market Share Analysis, 2018-2019 6.7 Growth Share Analysis

7 Global Minimal Residual Disease Testing Market (by Technology), 2019-2025 7.1 Overview 7.2 Flow Cytometry 7.3 Polymerase Chain Reaction (PCR) 7.4 Next-Generation Sequencing (NGS) 7.5 Other Technologies

8 Global Minimal Residual Testing Market (by Application), 2019-2025 8.1 Overview 8.2 Hematological Malignancies 8.2.1 Non-Hodgkin's Lymphoma (NHL) 8.2.2 Multiple Myeloma (MM) 8.2.3 Acute Lymphoblastic Leukemia (ALL) 8.2.4 Chronic Lymphocytic Leukemia (CLL) 8.2.5 Acute Myeloid Leukemia (AML) 8.2.6 Chronic Myeloid Leukemia (CML) 8.2.7 Hodgkin's Lymphoma (HL) 8.2.8 Other Leukemia

9 Global Minimal Residual Disease Testing Market (by End User), 2019-2025 9.1 Overview 9.2 Specialty Clinics and Hospitals 9.3 Diagnostic Laboratories 9.4 Research Institutions 9.5 Other End Users

10 Global Minimal Residual Disease Testing Market (by Region), 2019-2025 10.1 Overview

11 Company Profiles

For more information about this report visit https://www.researchandmarkets.com/r/qw8u11

Research and Markets also offers Custom Research services providing focused, comprehensive and tailored research.

Read the original:
Global MRD (Minimal Residual Disease) Testing Market Report 2020-2025: One of the Most Rapidly Evolving and Dynamic Markets - GlobeNewswire

Dr. Teresa Cody Offers Solution For Health And Wellness In 2021 – Press Release – Digital Journal

Platelet Rich Plasma Able to Assist in Healing Multiple Issues

Dr. Teresa Cody, a Sugar Land dentist, owner of the C and C Wellness and author of You Healing You, offers a solution for continued good health and wellness for multiple issues in 2021.

The underlying miraculous golden healing liquid known as Platelet Rich Plasma (PRP) in the medical industry is Codys tool to help people overcome some of their most difficult injuries and wounds.

Cody explains how doctors use this knowledge to concentrate this priceless blood component and reintroduce it into your body to heal injuries or as an esthetic treatment. Readers can learn how your own body holds the key to healing. It truly is you healing you.

One example is a patient with Rotator Cuff Injury where range of motion improved after two injection with the patients own PRP according to Cody. In her blog, Cody detailed that there are many reports of a professional sports person using PRP on tendon or ligament injuries.

He planned on getting surgery in a few months, however he thought he would give PRP a try first. Two injections in his shoulder and within 10 minutes his pain was gone, and his range of motion improved dramatically. This is not the first time we have seen this type of reaction. My theory is that the muscles surrounding the injury relax allowing greater range of motion. Muscles tend to tense and not let go. I know the rotator cuff has not repaired itself in 10 minutes, wrote Cody.

Another example of the effectiveness of PRP Cody explained in a blog post was regarding a patient with Dupuytrens contracture. This is a condition wherein the tissue in the palm of the hand thickens like a stiff cable causing the fingers to bend toward the palm. It most often affects the 4th or ring finger and 5th or little finger.

The PRP results are phenomenal.

After injecting PRP into the thick tissue, 80 percent of the thickness softened within 2 days, said Cody in her blog. The best part is that PRP is not a pharmaceutical it is the healing factors inside of each of us. We draw a small amount of blood and run it in a centrifuge so that the heavier red blood cells fall to the bottom and the plasma floats on top. After the plasma is pipetted into a syringe, it is injected into the injured area concentrating the bodys own healing factors.

Codys book is available on Amazon at http://www.amazon.com/You-Healing-Teresa-Cody-ebook/dp/B08MWTBX8N.

For more information, go tohttps://candcwellness.com.

Media Contact Company Name: Abundantly Social Contact Person: Aimee Ravichandran Email: Send Email Phone: 210.452.3622 Country: United States Website: https://candcwellness.com/

Here is the original post:
Dr. Teresa Cody Offers Solution For Health And Wellness In 2021 - Press Release - Digital Journal

The 9 Best Hair Growth Products That Work, According to Dermatologists – PureWow

Can we all agree that 2020 was a stressful year? So perhaps it comes as no surprise that there has been an uptick in people reporting hair loss, which can be triggered by stress, among other things.

To shed some light on how to best treat shedding hairs, we spoke to two board-certified dermatologistsAnnie Chiu, who is the founder of The Derm Institute in Los Angeles and Tess Mauricio in Beverly Hills, and Dr. Sophia Kogan, co-founder and Chief Medical Advisor of Nutrafolas well as Jen Atkin, a celebrity hairstylist, for some advice.

For starters, youve got to try and relax as much as you can. Right now [due to COVID-19], we are living through a prolonged period of stressful events, so this type of stress-induced hair loss is occurring at a higher rate than usual, explains Chiu. Time almost always helps, but in the meantime, you can find ways to help you manage your stress, like journaling, aromatherapy, taking long baths, and drinking chamomile tea.

Kogan also recommends incorporating activities like reading a book, meditating, yoga and dance into your day. Stress can be a trigger for hair thinning in many people, particularly women who tend to be more sensitive to its effects. Incorporating stress reduction techniques into your routine can do wonders for your body, mind and hair health.

When you are experiencing telogen effluvium, or sudden hair loss due to physical or mental stress to your body, its important to supply it with a well-balanced diet, says Chiu. Iron and biotin in particularly are very important. I also like collagen, overall vitamins, as well as saw palmetto extract.

You should also check your shampoos and other styling products. Chiu recommends staying away from drying and harsh ingredients like denatured alcohol and heavy silicones that can cause breakage and weigh your hair down. And avoid heat-styling your hair and being too rough with it when brushing. Both can lead to more breakage, which amplifies the look of hair loss.

Another consideration from Atkin: Switch to using a silk pillowcase, because ordinary pillowcases (which are typically made of other fabrics like cotton) can cause your hair to pull and tangle while you sleep. Also, its important to care for your hair with weekly masks and trims every three or so months to keep the ends healthy and prevent any splitting.

The ingredients to look for can vary based on the specific needs of an individual, and I always recommend consulting with your physician before adding anything new to your routine, cautions Kogan. Given the proliferation of products available to us, "its important to note that not all vitamins and supplements are created equal, so you want to pay close attention to the sourcing, quality and dosage of the ingredients contained in the products you are ingesting," she adds.

With that said, Mauricio shared some ingredients that have been shown to help with hair health and growth:

What results can you realistically expect from taking hair growth vitamins or supplements?

Most people report that their ponytail is thicker than it was previously and that their hair is growing much faster, says Chiu. However, all of the experts we interviewed agree that there is no single miracle cure for hair thinning and loss and treating it is a long game that requires patience and consistency.

Any product that claims to cure hair loss overnight or in a number of weeks should be viewed with skepticism, adds Kogan. Supplements can support hair growth and help build healthier hair, but they cannot bring dead follicles back to life. Nothing can.

When we are young and healthy, hair follicles contain and produce multiple hairs at once. With age, hair quality and growth can change due to multiple factors, explains Kogan. In some people, hair follicles can shrink, go dormant, die and then be replaced. Some dormant follicles have potential for regrowth, but others do not. A board-certified dermatologist can help distinguish what type of hair disorder is present and what may help.

Bottom line: Healthy hair growth is a slow and steady process that can be supported by promoting wellness from within the body, which is where supplements and vitamins come in. On their own, they dont solve the issue of hair loss, but they can support growth by creating an optimal environment for hair health and by targeting the underlying causes of hair thinning such as stress, hormones, gut health, nutrition and other environmental factors.

Because of the hair cycle (on average, your hair grows up to one inch in two months), it may take a few months before you see results from taking hair supplements, says Mauricio. There is no instant gratification. You have to be dedicated and patient.

The exact timeline varies from person to person, but ideally youll see results within six months, says Chiu, at which point youll notice more baby hairs coming in and your scalp will be less visible.

These supplements are best for people experiencing sudden hair loss due to a temporary shock to their body, whether its from stress, illness (like a bad cold or flu), or post-partum. If youre experiencing hair loss due to a more serious issue, supplements might help but its best to consult your physician first.

If you have any food allergies, I would take caution, says Chiu. For some people, biotin supplements can lead to acne. Also, if youre getting bloodwork done for anything, let your physician know that you are currently taking biotin as it can interfere with certain lab tests, she adds. Depending on the test, your physician may ask that you stop to ensure accurate results.

Kogan, who is the co-founder and Chief Medical Advisor of Nutrafol (a hair supplement), cautions that it's for adult use only and also recommends that pregnant or breastfeeding women refrain from taking [their] supplements. We likewise recommend that anyone on medications (especially blood thinners) or with medical conditions check with their primary care physician before starting a new supplement regimen.

Mauricio agrees, adding that because there are many reasons for hair loss and thinning, which can include underlying medical conditions, its important to consult with your doctor because treating the underlying condition can result in the reversal of hair loss altogether.

Topical scalp serums like Foligains Triple Action Hair Total Solution can help stimulate follicles to help with hair growth, says Chiu. And if seeing a board-certified dermatologist is an option, Platelet-Rich Plasma (PRP) injections can be effective for many types of hair loss.

Luckily, this is a growing field. We now have many more potential treatments for hair loss than ever before, says Mauricio. In addition to nutritional supplements, there are prescription medications like Finasteride, topical treatments like Rogaine and exosomes, at-home laser devices, and regenerative therapies like the use of patients own growth factors from platelet-rich plasma, platelet-rich fibrin matrix, and fat-derived stem cells. When used in combination, you can get the best results.

Ready to shop some expert picks ahead?

See the original post here:
The 9 Best Hair Growth Products That Work, According to Dermatologists - PureWow

Hemostemix Announces the Bread Contract with the Department of Foreign Affairs, Trade & Development Canada – BioSpace

Calgary, Alberta--(Newsfile Corp. - January 22, 2021) - .Hemostemix Inc (TSXV: HEM) (OTC: HMTXD) ("Hemostemix" or the "Company") is pleased to announce it has signed the Building Relationships Entrepreneurs & Dealmakers (BREAD) contract with the Department of Foreign Affairs, Trade and Development. An initiative to assist high-potential, biotech focused Canadian Small and Medium Enterprise (SMEs), the program is designed to accelerate the growth of Hemostemix and other Canadian biotechnology companies.

"We are actively working with the Trade Commissioner Service of CANADA in the USA, Japan and South Korea to source qualified partners to go to market with," stated Thomas Smeenk, CEO. "The BREAD agreement marks our Company's starting point to out-license ACP-01, and it generates our sponsorship into BioCom."

ABOUT THE TRADE COMMISSIONER SERVICE OF CANADA

The Trade Commissioner Service (TCS) plays an active role in helping Canadian companies achieve their goals of growth into international markets. Its services focus on helping companies prepare for international markets, assessing market potential, finding qualified contacts and partners and resolving problems.

ABOUT HEMOSTEMIX

Hemostemix is a publicly traded autologous stem cell therapy company, founded in 2003. A winner of the World Economic Forum Technology Pioneer Award, the Company developed and is commercializing its lead product ACP-01 for the treatment of CLI, PAD, Angina, Ischemic Cardiomyopathy, Dilated Cardiomyopathy and other conditions of ischemia. ACP-01 has been used to treat over 300 patients, and it is the subject of a randomized, placebo-controlled, double blind trial of its safety and efficacy in patients with advanced critical limb ischemia who have exhausted all other options to save their limb from amputation.

On October 21, 2019, the Company announced the results from its Phase II CLI trial abstract presentation entitled "Autologous Stem Cell Treatment for CLI Patients with No Revascularization Options: An Update of the Hemostemix ACP-01 Trial With 4.5 Year Follow-up", which noted healing of ulcers and resolution of ischemic rest pain occurred in 83% of patients, with outcomes maintained for up to 4.5 years.

The Company owns 91 patents across five patent families titled: Regulating Stem Cells, In Vitro Techniques for use with Stem Cells, Production from Blood of Cells of Neural Lineage, and Automated Cell Therapy. For more information, please visitwww.hemostemix.com.

For further information, please contact:

Thomas Smeenk, President, CEO & Co-Founder Suite 1150, 707 - 7thAvenue S.W., Calgary, Alberta T2P 3H6 Phone: 905-580-4170

Neither the TSX Venture Exchange nor its Regulation Service Provider (as that term is defined under the policies of the TSX Venture Exchange) accepts responsibility for the adequacy or accuracy of this news release.

Forward-Looking Information: This news release contains "forward-looking information" within the meaning of applicable Canadian securities legislation. All statements, other than statements of historical fact, included herein are forward-looking information. In particular, this news release contains forward-looking information in relation to: the commercialization of ACP-01. There can be no assurance that such forward-looking information will prove to be accurate. Actual results and future events could differ materially from those anticipated in such forward-looking information. This forward-looking information reflects Hemostemix's current beliefs and is based on information currently available to Hemostemix and on assumptions Hemostemix believes are reasonable. These assumptions include, but are not limited to: the underlying value of Hemostemix and its common shares; the successful resolution of the litigation that Hemostemix is pursuing or defending (the "Litigation"); the results of ACP-01 research, trials studies and analysis, including the midpoint analysis, being equivalent to or better than previous research, trials or studies as well as management's expectations of anticipated results; Hemostemix's general and administrative costs remaining constant; the receipt of all required regulatory approvals for research, trials or studies; the level of activity, market acceptance and market trends in the healthcare sector; the economy generally; consumer interest in Hemostemix's services and products; competition and Hemostemix's competitive advantages; and Hemostemix obtaining satisfactory financing to fund Hemostemix's operations including any research, trials or studies, and the Litigation. Forward-looking information is subject to known and unknown risks, uncertainties and other factors that may cause the actual results, level of activity, performance or achievements of Hemostemix to be materially different from those expressed or implied by such forward-looking information. Such risks and other factors may include, but are not limited to: the ability of Hemostemix to complete its current CLI clinical trial, complete a satisfactory futility analysis and the results of such and future clinical trials; litigation and potential litigation that Hemostemix may face; general business, economic, competitive, political and social uncertainties; general capital market conditions and market prices for securities; delay or failure to receive board or regulatory approvals; the actual results of future operations including the actual results of future research, trials or studies; competition; changes in legislation affecting Hemostemix; the timing and availability of external financing on acceptable terms; long-term capital requirements and future developments in Hemostemix's markets and the markets in which it expects to compete; lack of qualified, skilled labour or loss of key individuals; and risks related to the COVID-19 pandemic including various recommendations, orders and measures of governmental authorities to try to limit the pandemic, including travel restrictions, border closures, non-essential business closures, service disruptions, quarantines, self-isolations, shelters-in-place and social distancing, disruptions to markets, disruptions to economic activity and financings, disruptions to supply chains and sales channels, and a deterioration of general economic conditions including a possible national or global recession or depression; the potential impact that the COVID-19 pandemic may have on Hemostemix may include a decreased demand for the services that Hemostemix offers; and a deterioration of financial markets that could limit Hemostemix's ability to obtain external financing. A description of additional risk factors that may cause actual results to differ materially from forward-looking information can be found in Hemostemix's disclosure documents on the SEDAR website atwww.sedar.com. Although Hemostemix has attempted to identify important factors that could cause actual results to differ materially from those contained in forward-looking information, there may be other factors that cause results not to be as anticipated, estimated or intended. Readers are cautioned that the foregoing list of factors is not exhaustive. Readers are further cautioned not to place undue reliance on forward-looking information as there can be no assurance that the plans, intentions or expectations upon which they are placed will occur. Forward-looking information contained in this news release is expressly qualified by this cautionary statement. The forward-looking information contained in this news release represents the expectations of Hemostemix as of the date of this news release and, accordingly, it is subject to change after such date. However, Hemostemix expressly disclaims any intention or obligation to update or revise any forward-looking information, whether as a result of new information, future events or otherwise, except as expressly required by applicable securities law.

To view the source version of this press release, please visithttps://www.newsfilecorp.com/release/72606

Continue reading here:
Hemostemix Announces the Bread Contract with the Department of Foreign Affairs, Trade & Development Canada - BioSpace

Comprehensive Report on Stem Cell Banking Market 2021 | Trends, Growth Demand, Opportunities & Forecast To 2027 |Cordlife, Cryo-Cell…

Stem Cell Banking Market research report is the new statistical data source added by A2Z Market Research.

Stem Cell Banking Market is growing at a High CAGR during the forecast period 2021-2027. The increasing interest of the individuals in this industry is that the major reason for the expansion of this market.

Stem Cell Banking Market research is an intelligence report with meticulous efforts undertaken to study the right and valuable information. The data which has been looked upon is done considering both, the existing top players and the upcoming competitors. Business strategies of the key players and the new entering market industries are studied in detail. Well explained SWOT analysis, revenue share and contact information are shared in this report analysis.

Get the PDF Sample Copy (Including FULL TOC, Graphs and Tables) of this report @:

https://www.a2zmarketresearch.com/sample?reportId=64796

Note In order to provide more accurate market forecast, all our reports will be updated before delivery by considering the impact of COVID-19.

Top Key Players Profiled in this report are:

Cordlife, Cryo-Cell International, Cryo-Save Ag (A Subsidiary Of Esperite N.V), Lifecell International, Stemcyte, Viacord (A Subsidiary Of Perkinelmer), Global Cord Blood, Smart Cells International, Vita34, Cryoholdco.

The key questions answered in this report:

Various factors are responsible for the markets growth trajectory, which are studied at length in the report. In addition, the report lists down the restraints that are posing threat to the global Stem Cell Banking market. It also gauges the bargaining power of suppliers and buyers, threat from new entrants and product substitute, and the degree of competition prevailing in the market. The influence of the latest government guidelines is also analyzed in detail in the report. It studies the Stem Cell Banking markets trajectory between forecast periods.

Regions Covered in the Global Stem Cell Banking Market Report 2021: The Middle East and Africa (GCC Countries and Egypt) North America (the United States, Mexico, and Canada) South America (Brazil etc.) Europe (Turkey, Germany, Russia UK, Italy, France, etc.) Asia-Pacific (Vietnam, China, Malaysia, Japan, Philippines, Korea, Thailand, India, Indonesia, and Australia)

Get up to 30% Discount on this Premium Report @:

https://www.a2zmarketresearch.com/discount?reportId=64796

The cost analysis of the Global Stem Cell Banking Market has been performed while keeping in view manufacturing expenses, labor cost, and raw materials and their market concentration rate, suppliers, and price trend. Other factors such as Supply chain, downstream buyers, and sourcing strategy have been assessed to provide a complete and in-depth view of the market. Buyers of the report will also be exposed to a study on market positioning with factors such as target client, brand strategy, and price strategy taken into consideration.

The report provides insights on the following pointers:

Market Penetration: Comprehensive information on the product portfolios of the top players in the Stem Cell Banking market.

Product Development/Innovation: Detailed insights on the upcoming technologies, R&D activities, and product launches in the market.

Competitive Assessment: In-depth assessment of the market strategies, geographic and business segments of the leading players in the market.

Market Development: Comprehensive information about emerging markets. This report analyzes the market for various segments across geographies.

Market Diversification: Exhaustive information about new products, untapped geographies, recent developments, and investments in the Stem Cell Banking market.

Table of Contents

Global Stem Cell Banking Market Research Report 2021 2027

Chapter 1 Stem Cell Banking Market Overview

Chapter 2 Global Economic Impact on Industry

Chapter 3 Global Market Competition by Manufacturers

Chapter 4 Global Production, Revenue (Value) by Region

Chapter 5 Global Supply (Production), Consumption, Export, Import by Regions

Chapter 6 Global Production, Revenue (Value), Price Trend by Type

Chapter 7 Global Market Analysis by Application

Chapter 8 Manufacturing Cost Analysis

Chapter 9 Industrial Chain, Sourcing Strategy and Downstream Buyers

Chapter 10 Marketing Strategy Analysis, Distributors/Traders

Chapter 11 Market Effect Factors Analysis

Chapter 12 Global Stem Cell Banking Market Forecast

Buy Exclusive Report @:

https://www.a2zmarketresearch.com/buy?reportId=64796

If you have any special requirements, please let us know and we will offer you the report as you want.

About A2Z Market Research:

The A2Z Market Research library provides syndication reports from market researchers around the world. Ready-to-buy syndication Market research studies will help you find the most relevant business intelligence.

Our Research Analyst Provides business insights and market research reports for large and small businesses.

The company helps clients build business policies and grow in that market area. A2Z Market Research is not only interested in industry reports dealing with telecommunications, healthcare, pharmaceuticals, financial services, energy, technology, real estate, logistics, F & B, media, etc. but also your company data, country profiles, trends, information and analysis on the sector of your interest.

Contact Us:

Roger Smith

1887 WHITNEY MESA DR HENDERSON, NV 89014

[emailprotected]

+1 775 237 4147

https://neighborwebsj.com/

View original post here:
Comprehensive Report on Stem Cell Banking Market 2021 | Trends, Growth Demand, Opportunities & Forecast To 2027 |Cordlife, Cryo-Cell...

The chromosomal protein SMCHD1 regulates DNA methylation and the 2c-like state of embryonic stem cells by antagonizing TET proteins – Science Advances

RESULTS Identification of SMCHD1 as a protein associated with TET proteins

Using mass spectrometry (MS), we identified SMCHD1 as a protein interacting with FLAG-tagged TET3 in 293T cells (Fig. 1, A and B, and table S1), where it scored among the eight most significantly enriched proteins, which included TET3 itself and the known TET3 binding partner O-linked -N-acetylglucosamine transferase (OGT) (2729). To verify the SMCHD1-TET interaction further, we created ES cell lines by homologous recombination that carried a FLAG tag at the C terminus of the endogenous Smchd1 coding sequence (fig. S1A). We carried out anti-FLAG pulldown with one of the clones and performed proteomics analysis. Among the identified proteins were SMCHD1 itself as the highest-scoring protein (54% coverage) (fig. S1B and table S2). We detected the known SMCHD1-interacting protein, LRIF1 (12.3% coverage) (30). There were also several components of the PRC2 complex (EZH2, SUZ12, and MTF2; 4.5 to 15% coverage) but no PRC1 components. SMCHD1 has been shown to be a protecting factor against formation of histone H3 lysine 27 trimethylation (H3K27me3) by the Polycomb complex (25). In this proteomics experiment, we specifically recovered the TET2 protein (4.92% coverage), as associated with SMCHD1, but not TET1. TET3 is expressed at very low levels in ES cells. The known TET-interacting protein OGT (2729) was also identified (9.37%). We then transfected FLAG-tagged TET2 into 293T cells. Although TET2 itself was identified at only 4.0% coverage (fig. S1C and table S2), we still found SMCHD1 (11.4% coverage) and OGT (46%) as TET2-interacting proteins.

(A) Flag purification of TET3FL and TET3S from 293T cells. The purified samples were subjected to Coomassie blue staining (left) and Western blotting (right). The gel segments indicated were analyzed by MS. M, molecular weight markers; IB, immunoblot. (B) Identification of SMCHD1 as a binding partner of TET3 by MS. TET3S was expressed in 293T cells and immunoprecipitated with anti-FLAG beads. Gel segment 2 (A) was subjected to LC-MS/MS (liquid chromatographytandem MS) analysis (see Materials and Methods and table S1). The top eight highest-scoring proteins are shown. M.W., molecular weight. (C) Endogenous coimmunoprecipitation (co-IP) of SMCHD1 with TET1, TET2, and TET3FL. (D) Interaction between TET proteins and SMCHD1 by co-IP using expression of tagged proteins in 293T cells. (E) Different domains of TET3 were cotransfected with full-length SMCHD1 into 293T cells. After IP, the interacting proteins were identified by Western blotting. Stars indicate IgG (immunoglobulin G) bands. aa, amino acids. (F) Different domains of SMCHD1 were cotransfected with TET3FL into 293T cells. After IP, the interacting proteins were identified by Western blotting. Stars indicate IgG bands. HATPase, histidine kinase-like ATPase domain.

Using coimmunoprecipitation (co-IP), we found that all mouse TET proteins, including the long and short isoforms of TET3 [TET3FL (full-length TET3) and TET3S, respectively (31)], and TET1 and TET2 interact with SMCHD1 as endogenous proteins in cell types in which the proteins are expressed at substantial levels (TET1 and TET2 in ES cells and TET3 in Neuro2a cells) (Fig. 1C). These interactions were confirmed by cotransfection of the respective expression constructs and IP experiments (Fig. 1D).

To further substantiate the SMCHD1-TET interactions in cells, we performed bimolecular fluorescence complementation (BiFC) experiments (32, 33) with SMCHD1 and TET3 (fig. S2). The data suggest an efficient interaction of TET3 and SMCHD1 at a level similar to a positive control (P.C.) experiment with TET3 and OGT. The latter two proteins are interaction partners, as reported previously (27, 28). Although this assay does not determine that the two proteins interact directly, it is an independent confirmation of the in vivo TET-SMCHD1 interaction, for example, within a protein complex.

We then determined the interacting domains of TET3 and SMCHD1 by cotransfection experiments. We found that the C-terminal double-stranded -helix domain of TET3, which represents the core catalytic region conserved between all three TET proteins (8, 20, 34), interacts strongly with SMCHD1 (Fig. 1E). Analyzing SMCHD1 domains, we found that the N-terminal region including the GHKL adenosine triphosphatase (ATPase) domain interacted most efficiently with TET3FL (fragment F1; Fig. 1F), but the long central domain and the C-terminal hinge domain did not show any appreciable binding.

We then examined whether SMCHD1 and TET proteins can interact directly in vitro. We initially failed to observe a direct interaction between the recombinant N-terminal (ATPase) domain of SMCHD1 and the TET2 catalytic domain (TET2-CD). Next, we prepared recombinant full-length SMCHD1 and recombinant TET2-CD or TETFL proteins. These proteins were purified from either baculovirus-infected cells (SMCHD1 and TET2-CD) or from mammalian cells (TET2FL or TET1FL). We used the AlphaScreen system (fig. S3A) for assessing binding reactions in a quantitative biophysical assay. In these assays, SMCHD1-FLAG was biotinylated via an introduced C-terminal biotinylation sequence (AviTag) and was coexpressed along with biotin ligase (BirA) in baculovirus-infected insect cells. This SMCHD1 protein could be collected on streptavidin beads indicating that it was biotinylated successfully (fig. S3B). The mammalian TET proteins were expressed as C-terminal His-tagged proteins (fig. S3B). In the AlphaScreen assays, binding between a biotinylated protein captured on streptavidin AlphaScreen donor beads and a His-tagged protein captured on nickel chelate (Ninitrilotriacetic acid) AlphaScreen acceptor beads is measured. After performing these assays, we did not observe any direct binding between SMCHD1 and TET2 proteins (TET1 tested negatively as well) (fig. S3C). We also performed in vitro biotinylation of SMCHD1 using biotinylation kits, or we biotinylated the anti-SMCHD1 antibody followed by SMCHD1 binding. We concluded that our consistently observed interactions between SMCHD1 and TET proteins in cells are most likely based on an indirect interaction, perhaps involving a larger protein complex or a bridging protein. One potential candidate for such a protein is OGT, which we recovered in SMCHD1 and TET protein complexes and which is known to interact with the TET2/3-CDs (27, 29, 35, 36).

When SMCHD1 was coexpressed in 293T cells together with TET3FL, TET activity was inhibited in an SMCHD1 dose-dependent manner, leading to the formation of lower amounts of the TET reaction product 5hmC (Fig. 2A), while total levels of 5mC did not change appreciably. Another in vivo assay of TET activity is based on demethylation and reactivation of a luciferase vector that is methylated in vitro at all CpG sites before transfection. In this assay, TET3S is more active than TET3FL (31). The luciferase activity of the fully CpG-methylated luciferase reporter vector was increased by cotransfection of TET3S, as reported previously (Fig. 2B) (31). This TET-induced activity was inhibited by SMCHD1, which did not reduce the activity of an unmethylated control reporter (Fig. 2B).

(A) Reduction in 5hmC levels by coexpression of SMCHD1 with TET3 in 293T cells. 5hmC and 5mC contents were assessed using antibody-based dot blots. One-way analysis of variance (ANOVA) was performed comparing the mean of each group with the mean of the second group (**P < 0.01 and ***P < 0.001; mean SEM). ns, not significant. (B) Inhibition of TET3S-induced reactivation of a methylation-silenced luciferase construct by SMCHD1 in 293T cells (top). One-way ANOVA was performed (**P < 0.01 and ****P < 0.0001). Data are for means SEM of three independent experiments. An unmethylated luciferase vector was used as a control (bottom). (C) FLAG purification of TET2-CD and SMCHD1 full length (SMCHD1-FL) from Sf9 insect cells. Coomassie blue staining. (D) Inhibition of TET2-CD activity on fully methylated DNA in the presence of SMCHD1 as shown by combined bisulfite restriction analysis (COBRA) assay (BstU I cleavage indicates methylation). P.C., positive control with excess TET protein (18 g); N.C., negative control without TET treatment. Different molar ratios of SMCHD1 and TET protein (1.15 g) are shown. The H19 imprinting control region was analyzed. (E) Bisulfite sequencing analysis of H19 methylation analyzed in duplicates. Solid black circles indicate modified CpGs; open circles indicate TET-oxidized mCpGs. The purple arrows indicate BstU I sites. (F) Percentages of modified cytosines (%Me) of the different samples. P values were determined by Fishers exact test (two sided).

We then proceeded to purify recombinant active TET proteins (TET2-CD and TET2FL) and full-length SMCHD1 from baculovirus-infected cells (Fig. 2C). TET2-CD was catalytically more active than TET2FL and was therefore used in our in vitro activity assays. The in vitro activity of TET2 was initially tested using combined bisulfite restriction analysis (COBRA) (37), an assay in which cleavage with BstU I (5CGCG) indicates methylation at those sites. In this assay, addition of SMCHD1 inhibited TET activity in a dose-dependent manner (Fig. 2D). We further verified this effect by sodium bisulfite sequencing (Fig. 2, E and F). This assay monitors the end-product of TET activity, 5caC, which scores as unmodified cytosine in bisulfite sequencing due to decarboxylation and deamination of 5caC. SMCHD1 inhibited TET activity effectively at a 1:1 molar ratio and caused almost complete inhibition at a ratio of 2:1 (Fig. 2E). SMCHD1 is a DNA-binding protein with binding likely mediated through its hinge domain (3840). We propose that binding of SMCHD1 to DNA leads to an occlusion of TET activity from its DNA target.

Next, we created and verified several Smchd1 knockout (KO) clones of male mouse ES cells (mESCs) using CRISPR-Cas9 technology (Fig. 3A and fig. S4A). Using immuno-dot blots, we determined that these KO clones have moderately increased levels of 5hmC (fig. S4B), suggesting that the lack of SMCHD1 leads to stimulation of the 5mC oxidation process, which is consistent with the in vitro data showing that SMCHD1 inhibits TET activity. Globally, TET and DNMT protein expression was not significantly altered in SMCHD1-deficient cells (fig. S4, C and D).

(A) Absence of SMCHD1 protein in three CRISPR-Cas9 KO ES cell clones. (B) Heatmap of RNA-seq data indicates differentially expressed genes between WT (n = 3 clones) and SMCHD1 KO (n = 3 clones) ES cells. (C) Gene set enrichment analysis (GSEA) of the 2c-like ES cell signature. The gene set represents genes activated during zygotic genome activation in 2c mouse embryos and enriched in 2C::tomato+ cells (42). The x axis shows the log2 fold change of the KO/WT-ranked transcriptome. GSEA analysis was performed as previously described (49). (D) The heatmap indicates the differentially expressed 2c-like genes between WT (n = 6) and SMCHD1 KO (n = 6) ES cells including two technical replicates for each clone. Typical 2c-like genes, such as Dux (indicated by red arrow), Zscan4c, Dub1, and Usp17l family members (indicated by purple arrows) are indicated. (E) The density plot indicates activation of repeat elements in SMCHD1 KO cells. The x axis shows the log2 (fold change of KO/WT) of repeat element expression. The y axis shows the density.

Next, we performed whole-genome bisulfite sequencing (WGBS) on three wild-type (WT) and three Smchd1 KO ES cell clones. We observed lower levels of modified cytosines in the knockouts on all chromosomes except for the Y chromosome, which became hypermethylated (fig. S5A). This moderate global reduction of modified cytosines affected all genomic compartments except for CpG islands, which have constitutively very low levels of methylation (fig. S5B). Using DMRseq analysis (41), we identified 283 hypomethylated and 223 hypermethylated differentially methylated regions (DMRs) in the clones lacking SMCHD1 (fig. S5C and table S3). One extensively hypomethylated genomic region was the Pcdha gene cluster, which is a known binding region for SMCHD1 (40).

To look for functionally relevant epigenetic changes upon loss of SMCHD1, we performed RNA sequencing (RNA-seq) and identified 1236 up-regulated and 256 down-regulated genes [fold change > 2, false discovery rate (FDR) < 0.05] in the Smchd1 KO cells compared to WT cells (Fig. 3B and table S3). This is consistent with a role of SMCHD1 as a transcriptional repressor. Up-regulated genes, but not down-regulated or unchanged genes, had slightly reduced levels of DNA methylation near the transcription start sites (TSSs) (fig. S5, D to F). There was an overall negative correlation between the direction of methylation change in the SMCHD1 knockouts and the expression change of the DMR-associated genes (fig. S3G). Gene ontology analysis for DMR-associated differentially expressed genes pointed to an enrichment of pattern-specific and organ developmentspecific processes (fig. S3H).

Using gene set enrichment analysis (GSEA), one notably up-regulated set of genes was identified as 2c embryolike genes (Fig. 3, C and D), which reflected the up-regulation of 136 genes normally expressed in 2c mouse embryos as a feature of the initial wave of zygotic genome activation (ZGA) (42). A similar set of genes found in ZSCAN4+ mESCs (43) or in CAF1 knockdown ES cells (44) was also enriched in the group of genes up-regulated in SMCHD1 KO cells. A small percentage of WT ES cells sporadically express 2c embryo stagespecific (2C) transcripts, such as Zscan4, and cycle in and out of this specialized state (42). Among the genes up-regulated in the absence of SMCHD1 was the Zscan4 gene cluster (Fig. 4A). The up-regulation of Zscan4, which encodes a protein involved in telomere maintenance (45), was confirmed at the protein level using a pan-ZSCAN4 antibody (Fig. 4B). The fraction of ZSCAN4-positive cells in the ES cell population increased from ~1.5% in WT cells to about 12% in the SMCHD1 KO cells (Fig. 4, C and D). Various repetitive element families, such as murine endogenous retroviruses (MERVK and MERVL), which are activated by ZSCAN4 (46) and also derepressed during ZGA (42), showed increased expression upon loss of SMCHD1 (Fig. 3E).

(A) Integrative Genomics Viewer screenshots of RNA-seq track peaks across all Zscan4 family members in WT and SMCHD1 KO ES cells. (B) Up-regulation of ZSCAN4 protein in SMCHD1 KO cell lines. (C) The fraction of ZSCAN4-positive cells in the ES cell population is increased in the absence of SMCHD1. ES cells were immunostained for ZSCAN4 (green). DNA was counterstained with DAPI (4,6-diamidino-2-phenylindole) (blue). Scale bars, 50 m. (D) Fractions of ZSCAN4+ cells in WT ES cells and Smchd1-KO ES cells. t test was performed for statistical analysis (P < 0.001). Error bars indicate SEM (six independent experiments). (E) Browser view of RNA-seq tracks across the Dux locus in WT and SMCHD1 KO ES cells. The Dux gene itself is shaded in yellow. (F) Quantitative real-time polymerase chain reaction (qRT-PCR) data confirm Dux activation upon SMCHD1 loss. -Actin was used as a control. One-way ANOVA was performed for statistical analysis, comparing the mean of each group with the mean of the WT group (***P < 0.001). Data are for means SEM of three independent KO clones.

We then focused our attention on the Dux locus, which encodes a double-homeobox transcription factor (DUX) implicated in ZGA and in 2c-like transcriptomes (4749). We found that Dux is strongly (>10-fold) activated in SMCHD1-deficient ES cells (Fig. 4, E and F). Within the same locus, several other transcripts including the 5UTR (5 untranslated region) of a Dux pseudogene (Gm4981) were also up-regulated upon loss of SMCHD1 (Fig. 4E). WGBS (Fig. 5A) and manual bisulfite sequencing (Fig. 5B) revealed substantial demethylation of the Dux promoter in the SMCHD1 KO clones (WGBS, P < 1 105; manual bisulfite sequencing, P < 0.01; t test). We then incorporated a MERVL-promoter-dTomato reporter construct into the SMCHD1 KO and WT ES cells and purified dTomato-expressing (2c-like) cells by fluorescence-activated cell sorting (FACS) (Fig. 5C). In this cell population, methylation of the Dux promoter was even further reduced compared to unsorted cell populations, and methylation levels were lowest in the sorted SMCHD1 KO cells (Fig. 5, D and E). Using chromatin IP (ChIP) and quantitative polymerase chain reaction (qPCR), we observed that the SMCHD1 protein is present at the two different locations examined in the mouse Dux promoter in WT, but not in SMCHD1 KO ES cells (fig. S6A).

(A) Single CpG modification levels of WT and SMCHD1 KO samples at the Dux locus, as determined by WGBS. The differential methylation region is shaded in purple. CpGs are denoted with tick marks. Red circles, WT; blue circles, SMCHD1 KO. Circle size is proportional to coverage. A smoothed line is shown for each sample. (B) Manual bisulfite sequencing of the Dux promoter in WT and SMCHD1 KO cells. Solid black circles indicate modified CpG sites; open circles indicate unmodified CpG sites. Total percentages of modified cytosines (%Me) are shown. The primers are also indicated in (A). (C) Representative fluorescence image and FACS plot of 2C::tdTomato+ cells in the Smchd1 KO ES cell populations. (D) Methylation of the Dux promoter in the dTomato-expressing (FACS-sorted) Smchd1 KO cell population is further reduced. The data show bisulfite sequencing analysis of the Dux promoter in reporter-expressing WT and SMCHD1 KO ES cells. Total percentages of modified cytosines (%Me) are shown. (E) Percentages of modified cytosines at the Dux promoter determined from (B) and (D). One-way ANOVA was performed (*P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001). Error bars indicate SEM from triplicate clones (WT and Smchd1-KO) or duplicate samples (FACS-sorted 2c-Smchd1-KO cells).

To examine whether TET-mediated 5mC oxidation is involved in demethylation of the Dux promoter when SMCHD1 is not functional, we analyzed 5hmC by a pulldown method following derivatization of the hydroxymethyl group with biotin (fig. S6B) (50) or by single-base resolution TET-assisted bisulfite sequencing [TAB sequencing; (51)] (fig. S6C; see fig. S7 for complete results). Both methods indicated a significant increase in 5hmC at the Dux promoter in SMCHD1-deficient cells. Using ChIP, we observed enhanced binding of TET1 to the R2 region of the Dux promoter in SMCHD1 KO cells (fig. S6D). This is the same location where SMCHD1 is normally bound (fig. S6A) in WT ES cells, suggesting a shielding effect of SMCHD1 toward the 5mC oxidase.

To delineate the contribution of Dux to the entire set of genes up-regulated after loss of SMCHD1, we biallelically inactivated Dux in Smchd1 KO ES cells and also in WT ES cells (confirmed by extensive DNA sequencing because no reliable anti-DUX antibody was available; Fig. 6A). The subsequent RNA-seq analysis showed that 47 of the 136 up-regulated 2c-like genes in SMCHD1 KO cells were no longer up-regulated in the absence of a functional Dux gene (Fig. 6, B and C). One example of such Dux-dependent genes is the Zscan4 gene cluster, the expression of which was strictly dependent on WT Dux (Fig. 6D). In a published study (48), 5738 genes were linked to HA-DUX peaks. On the basis of this gene set, we found that still, 49 of the 2c-like genes that are up-regulated in our Smchd1 single KO were not bound by DUX. So, we can confirm that a fraction of up-regulated 2c-like genes (49 of 136) in Smchd1-KO cells may not be directly regulated by DUX. The data indicate that DUX is a substantial but not exclusive contributor to the 2c-like transcriptome induced in the absence of SMCHD1 (Fig. 6).

(A) The small guide RNA (gRNA) targeting region is immediately downstream of the start codon (ATG) of the Dux gene. Sanger sequencing confirmed frameshift mutations. Sequences targeted by the gRNA are in blue, and the PAM (protospacer adjacent motif) sequence is shown in red. Biallelic frameshift mutation was shown for each clone. The gRNA was applied in WT and in SMCHD1 KO ES cells to obtain the Dux single-knockout and Smchd1/Dux double-knockout ES cell clones, respectively. (B) A heatmap indicates the differentially expressed 2c-like genes between Dux/Smchd1 double-knockout (n = 3 clones) and Smchd1 single-knockout (n = 3 clones, two replicates each) ES cells. The blue color indicates genes no longer up-regulated in the double knockouts. (C) The pie chart shows that 47 of the 136 single-KO up-regulated 2c-like genes were no longer up-regulated in the absence of Dux in the Smchd1/Dux double KO. (D) RNA-seq tracks generated by the GVIZ package across the Zscan4 gene family members in WT ES cells (black), Smchd1 KO ES cells (red), Dux KO ES cells (blue), and Smchd1 plus Dux (double-KO) ES cells (green).

Our data showed an interaction of SMCHD1 with TET proteins within cells (Fig. 1 and figs. S1 and S2) and an inhibition of TET-induced 5mC oxidation by SMCHD1 (Fig. 2). In the absence of SMCHD1, 5hmC levels are increased (fig. S4; fig. S6, B and C; and fig. S7), suggesting that SMCHD1 is a negative regulator of TET activities. To obtain genetic support for this interaction, we used Tet1/2/3 triple-knockout ES cells (52) and deleted SMCHD1 from these cells, as confirmed by Western blotting and DNA sequencing (Fig. 7A and fig. S8A). The Dux gene could no longer be activated upon loss of SMCHD1 in TET-TKO cells (Fig. 7, B and C; compare to Fig. 4, E and F). Consequently, DUX target genes such as Usp17lc, Zscan4f, and Zfp352 (4749), as well as other SMCHD1-regulated genes such as 1700013H16Rik and Gm12690, could also no longer be activated (Fig. 7D). Of the 136 2c-like genes up-regulated in the absence of SMCHD1, only 69 were still up-regulated in the absence of TET activities (Fig. 7E). In TET-TKO cells, the Dux promoter was >90% methylated at CpG sites. However, unlike in WT ES cells (Fig. 5B), loss of SMCHD1 in these cells did not elicit significant DNA demethylation (Fig. 7F). These experiments demonstrate that TET proteins are required to cause demethylation of the Dux promoter and activation of Dux when SMCHD1 is dysfunctional, genetically confirming that SMCHD1 operates as a negative regulator of TET activity.

(A) Absence of SMCHD1 protein in three CRISPR-Cas9targeted Tet triple-knockout ES cell clones. (B) RNA-seq tracks across Dux in WT, Tet triple-knockout ES cells, and Tet-Smchd1 quadruple-KO cells. (C) Quantitative RT-PCR analysis of Dux expression in WT, Tet triple-knockout and Tet-Smchd1 quadruple-knockout cells. One-way ANOVA was performed. Data are for means SEM of three independent clones. (D) RNA-seq analysis across different genes in WT, Smchd1 KO, Dux KO, Smchd1 and Dux (double) KO, Tet triple-knockout, and Tet-Smchd1 quadruple-knockout ES cells. One-way ANOVA was performed, comparing the mean of each group (n = 3 clones each) with the mean of the Smchd1-KO group (**P < 0.01 and ****P < 0.0001). Error bars indicate SEM. (E) The number of up-regulated 2c-like genes is decreased in the absence of TET proteins. (F) Bisulfite sequencing analysis of the Dux promoter in Tet-TKO cells and quadruple-knockout cells. Percentages of modified cytosines (%Me) are shown. (G) Eighty-nine percent (75 and 14%) of significantly up-regulated genes in the SMCHD1 single KO are no longer up-regulated in the absence of TET proteins in the Tet-Smchd1 quadruple-KO (qKO) cells. (H) Model of SMCHD1 as a negative regulator of TET proteins at the Dux promoter. Black circles, 5mC; light blue circles, 5hmC; white circles, unmethylated CpGs.

In the Smchd1, Tet1, Tet2, and Tet3 quadruple-knockout cells, a total of 921 of 1236 genes (75%), which were up-regulated in Smchd1 single-knockout cells, could no longer be activated (Fig. 7G and fig. S8B). Examples are shown in Fig. 7D. An additional 179 genes (14%), normally activated upon SMCHD1 loss, were even down-regulated in these quadruple knockouts (Fig. 7G). These data indicate that the aberrant transcriptome in the absence of SMCHD1 depends to a substantial extent on the presence of TET proteins (fig. S8, C and D).

In this study, we identified SMCHD1 as a TET-interacting protein initially by MS. There have been several previous studies in which TET- or SMCHD1-interacting proteins were analyzed by proteomics, but this interaction was not found (27, 28, 30, 53). One recent publication did identify SMCHD1 as a TET2-interacting protein in their proteomics data (54). Some studies used higher salt concentrations (300 mM) for cell lysis or washing steps (30, 53), but others used conditions similar to ours (27, 54). It is possible that the relatively milder extraction conditions we used may explain that we did find the TET-SMCHD1 association. The SMCHD1-TET complexes may be disrupted by 300 mM NaCl. Extraction and washing steps are certainly a determining factor for identification of interacting proteins. Using higher salt concentrations (>150 mM) will increase the risk of disassembling protein complexes. Other data in our manuscript, including endogenous co-IP, BiFC (which does not use cell extraction or salt washes), and the genetic studies, further support the TET-SMCHD1 interaction.

From our data, we propose a model in which SMCHD1 acts as a negative regulator of TET proteins by inhibiting their activity at target sequences (Fig. 7H). This regulation likely involves a shielding mechanism because DNA hypomethylation is most pronounced at known SMCHD1-bound genomic regions (e.g., Dux and Pcdha gene cluster). SMCHD1 may inhibit TET either by direct DNA binding or via its presence in heterochromatin. Localized TET inhibition or trapping of TET by SMCHD1 may also lead to a slight reduction in global 5mC oxidation, which is reversed upon SMCHD1 depletion leading to a moderate global DNA hypomethylation. Our data are conceptually consistent with other models that have posited that SMCHD1 functions in chromatin as an antagonistic protein against CCCTC binding factor (CTCF) binding (40), either by a shielding mechanism or by promoting DNA methylation that interferes with binding of CTCF. SMCHD1 also has been shown to be a protecting factor against formation of H3K27me3 by the Polycomb complex (25).

SMCHD1 loss-of-function mutation, often affecting its ATPase domain, is a hallmark of the human muscular dystrophy disease facioscapulohumeral dystrophy (FSHD2), which is characterized by inappropriate activation of the DUX4 gene, which is the human homolog of mouse Dux (5557). It is of interest to speculate that loss of TET restriction by dysfunction of SMCHD1 may lead to TET-induced hypomethylation of DUX4 control regions and unscheduled expression of DUX4, perhaps starting already during human embryonic development and later manifesting itself in muscle disease.

Our data suggest that SMCHD1 is critical for Dux suppression in mESCs, thus controlling the 2c-like state. Furthermore, SMCHD1 plays a key role in de novo methylation of CpG islands on the inactive X chromosome during mouse development (23). Recent data have shown, however, that loss of SMCHD1 in somatic cells does not lead to X chromosome reactivation (24, 25). Together, the existing data suggest that SMCHD1 functions in promoting de novo DNA methylation during development rather than in mediating methylation maintenance, and we propose the following mechanism: SMCHD1 operates in these critical DNA methylation events by inhibiting TET-mediated 5mC oxidation and demethylation at its target regions, as we show in this study, thereby shifting the balance of methylation versus demethylation toward the methylated state (Fig. 7H). When SMCHD1 is lost, but DNA methylation remains high in the absence of TETs (reduced DNA methylation dynamics), Dux expression may not be occurring because of inhibition of transcription by DNA methylation. On the other hand, in absence of SMCHD1 and presence of TET activity, and thus higher methylation-demethylation dynamics, Dux will be activated. This pathway is important in inhibiting the totipotent (2c-like state) of ES cells. Although a role of SMCHD1 in inactivation of Dux in late 2c mouse embryos has recently been proposed (58), this event seems initially independent of DNA methylation inasmuch as the Dux promoter region is almost completely unmethylated in 2c and 4c mouse embryos (59); therefore, the de novo methylation events must occur later during development. Further studies are needed to confirm whether SMCHD1 is responsible for the remethylation of the Dux locus during early embryo development and whether inhibition of TET proteins plays an important role in this process.

FLAG-tagged TET3FL or FLAG-tagged TET3S plasmids (31) were transfected into 293T cells. After 48 hours, cells were lysed in 10 mM tris-HCl (pH 7.4), 150 mM NaCl, 0.125% NP-40, and 2.5 mM EDTA. The lysate was added to M2 anti-FLAG affinity beads (Sigma-Aldrich), which were agitated overnight at 4C. After extensive washing with lysis buffer containing 200 mM NaCl followed by washing with 20 mM tris-HCl (pH 7.6) and 200 mM NaCl, the IP beads were mixed with 5 SDS loading buffer and heated for 10 min at 80C. Each protein sample was loaded onto 12% SDSpolyacrylamide gel electrophoresis (PAGE) gels. After visualization using Coomassie blue, the gel lanes were cut into eight segments and sliced into small pieces for in-gel digestion. Gel pieces were washed three times with distilled water to remove SDS and dehydrated using 100% acetonitrile. Proteins were treated with 10 mM dithiothreitol (DTT) in 50 mM NH4HCO3 for 45 min at 56C. After washing with 100% acetonitrile, alkylation of cysteines was performed with 55 mM iodoacetamide in 50 mM NH4HCO3 for 30 min in the dark. Last, each dehydrated gel piece was treated with sequencing-grade modified trypsin (12.5 ng/l; Promega, Madison, WI) in 50 mM NH4HCO3 buffer (pH 7.8) at 37C overnight. Following digestion, tryptic peptides were extracted with 5% formic acid in 50% acetonitrile solution at room temperature for 20 min. The supernatants were collected and dried in a SpeedVac. Samples were resuspended in 0.1% formic acid and were purified and concentrated using C18 ZipTips (Millipore, MA) before MS analysis.

Peptide separation was performed using a Dionex UltiMate 3000 RSLCnano system (Thermo Fisher Scientific). Tryptic peptides from bead columns were reconstituted using 0.1% formic acid and separated on a 50-cm EASY-Spray column with a 75-m inner diameter packed with 2-m C18 resin (Thermo Scientific, USA) over 120 min (300 nl/min) using a 0 to 45% acetonitrile gradient in 0.1% formic acid at 50C. The liquid chromatography (LC) was coupled to a Q Exactive Plus mass spectrometer with a nano-ESI source (Thermo Fisher Scientific). Mass spectra were acquired in a data-dependent mode with an automatic switch between a full scan with 10 data-dependent tandem MS (MS/MS) scans. Target value for the full-scan MS spectra was 3,000,000 with a maximum injection time of 120 ms and a resolution of 70,000 at mass/charge ratio (m/z) of 400. The ion target value for MS/MS was set to 1,000,000 with a maximum injection time of 120 ms and a resolution of 17,500 at m/z 400. Dynamic exclusion of repeated peptides was applied for 20 s.

The acquired MS/MS spectra were searched using SequestHT on Proteome Discoverer (version 2.2, Thermo Fisher Scientific) against the Swiss-Prot database. Briefly, precursor mass tolerance was set to 10 ppm (parts per million) and MS/MS tolerance was set at 0.02 Da. FDRs were set at 1% for each analysis using Percolator. From the Sequest search output, peptide data were default values of Proteome Discoverer. Label-free quantitation was performed using peak intensity for unique and razor peptides of each protein. Normalization was done using total peptide amount.

To identify TET2 interaction partners, we transfected FLAG-tagged TET2FL with N-terminal FLAG and HA tags (Addgene plasmid no. 41710; a gift from A. Rao) into 293T cells, harvested the cells after 48 hours, and processed them similar as described below for ES cells. ES cells in which the SMCHD1 protein was tagged endogenously with a C-terminal FLAG tag were prepared as follows: We followed the protocol of CETCh-seq (60). Briefly, we designed guide RNA (gRNA; 5GTCTTCAGAAATGCTCAGTT) and cloned it into pSpCas9-2A-puromycin (PX459, Addgene; a gift from F. Zhang) to target and cut near SMCHD1s stop codon. We cloned 700 to 800base pair(bp)long homology arms of Smchd1 into the pFETCh-donor backbone vector (Addgene plasmid no. 63934; gift from E. Mendenhall and R. M. Myers) by Gibson assembly reaction. Then, we cotransfected the donor plasmid and gRNA plasmid at a ratio of 2:1. The single FLAG-tagged cell clones were selected in puromycin (1.5 g/ml) and G418 (200 g/ml). The DNA of selected clones was extracted and sequenced to detect the presence of the FLAG tag. Tagged SMCHD1 protein was detected by Western blot with anti-FLAG antibody and anti-SMCHD1 antibody.

The cells were harvested and lysed in ice-cold lysis buffer consisting of 10 mM tris-HCl (pH 7.4), 150 mM NaCl, 0.125% (v/v) NP-40, cOmplete protease inhibitor tablets (Sigma-Aldrich; 1 tablet/10 ml), and 2.5 mM EDTA at 4C for 1 hour. We centrifuged the cell lysate at 40,000g for 60 min at 4C, then transferred the supernatant onto equilibrated anti-FLAG M2 affinity beads (Sigma-Aldrich), and incubated the slurry on a rotation wheel overnight at 4C. We washed the beads with ice-cold wash buffer containing 10 mM tris-HCl (pH 7.4), 250 mM NaCl, 0.125% (v/v) NP-40, cOmplete (1 tablet/10 ml), and 2.5 mM EDTA on a rotation wheel at 4C for 5 min and repeated the washing five times. Following the final wash, the beads were then eluted twice with elution buffer containing 20 mM tris-HCl (pH 7.5), 150 mM NaCl, 0.02% (v/v) Tween 20, and 3 FLAG peptide (150 g/ml) for 5 min. The eluted samples were then mixed with 5 SDS loading buffer and denatured for 10 min at 99C. The protein samples were loaded and separated on a mini gel (Bio-Rad Mini-PROTEAN TGX 4 to 20%). The gel was stained using Coomassie blue and destained water. The cut gel samples were digested with trypsin and injected into a Thermo Orbitrap Fusion Lumos mass spectrometer at the University of Massachusetts Proteomics Core Facility. The data were searched against the Swiss-Prot human/mouse database using the Mascot search engine through Proteome Discoverer software.

Full-length SMCHD1 was cloned into the pFastBac vector (Thermo Fisher Scientific) with a FLAG tag. We confirmed all expression vectors by Sanger sequencing. For FLAG-tagged SMCHD1 and TET2 protein expression, the bacmid DNA was transfected into Sf9 cells (Bac-to-Bac baculovirus expression system; Thermo Fisher Scientific) to obtain the passage 0 (P0) baculovirus at 96 hours after transfection. Then, we continued to generate P1 baculovirus by infecting the cells with P0 baculovirus. Proteins were expressed for 72 hours using 1000 ml of insect cells (2 million cells/ml) after transfecting P1 virus, and the cell pellet was resuspended in lysis buffer [50 mM Hepes (pH 7.5), 300 mM NaCl, 0.2% (v/v) NP-40, cOmplete, EDTA-free Protease Inhibitor Cocktail (Roche, 1 tablet/10 ml), and Benzonase Nuclease (10 U/ml) (Millipore) to destroy nucleic acids]. The lysate was cleared by centrifugation at 20,000g for 60 min. Anti-FLAG M2 affinity gel (Sigma-Aldrich) was equilibrated in lysis buffer following the manufacturers instructions. We incubated the cleared lysate with equilibrated FLAG M2 affinity gel at 4C for 2 hours. Bound protein was then washed five times with wash buffer [50 mM Hepes (pH 7.5), 150 mM NaCl, and 15% (v/v) glycerol]. We eluted the protein with the wash buffer containing 3 FLAG peptide (100 g/ml) (Sigma-Aldrich). Purified FLAG-tagged proteins were concentrated by Amicon Ultra Centrifugal Filters and DTT was added to 1 mM, then aliquots were flash-frozen in liquid nitrogen, and stored at 80C. The purification of TET proteins from mammalian cells was performed as described using anti-FLAG purification, as described above.

We performed TET protein in vitro assays on the basis of the established TET oxidation reaction (see TAB sequencing reactions for details). For a P.C., 18 g of TET2-CD protein was used to treat 500 ng of Sss Imethylated genomic DNA to get fully oxidized DNA containing 5caC. For a negative control (N.C.), no TET protein was used. For the testing samples, 1.15 g of TET protein was used to treat Sss Imethylated genomic DNA, and different amounts of recombinant full-length SMCHD1 were added into the TET oxidation reaction. Bovine serum albumin (BSA) was used as a control. For a blank control, only the elution buffer for protein purification was used to keep the volume of the reactions identical. After the TET oxidation reaction, we performed bisulfite conversion treatment on the purified DNA with the EZ DNA Methylation-Gold Kit (Zymo Research). This treatment converts the TET reaction product 5caC to uracil. For COBRA, BstU I was used to digest the PCR products obtained after bisulfite conversion. For sequence analysis, the PCR products obtained after bisulfite conversion were cloned into the Topo TA cloning vector, and clones were sequenced.

J1 mESCs (from American Type Culture Collection) were cultured under feeder-free conditions on 0.1% gelatincoated tissue culture plates in KO DMEM (Dulbeccos modified Eagles medium; Gibco, 10829-018) supplemented with 15% fetal bovine serum, LIF (1000 U/ml) (Millipore, ESG1106), 1 nonessential amino acids (Gibco, 11140-050), 100 M -mercaptoethanol (Invitrogen, 21985-203), and 2 mM l-glutamine (Gibco, 25030-081).

mESCs were transfected with pSpCas9-2A-puromycin (PX459) plasmids (Addgene plasmid no. 62988; a gift from F. Zhang) carrying the appropriate Smchd1 sgRNAs, by using the BioT transfection reagent (Bioland, B01-01) according to the manufacturers instructions. Single-cell clones were selected in puromycin (1.5 g/ml). To inactivate the Dux gene in WT and Smchd1 KO ES cell clones, we transfected WT cells or Smchd1 KO ES cells with a pSpCas9-2A-blasticidin plasmid carrying the appropriate Dux sgRNA. Single-cell clones were selected with blasticidin (8 g/ml). We used the same pSpCas9-2A-puromycin-gRNA vector to knock out Smchd1 in Tet1/Tet2/Tet3 triple-knockout ES cells. The DNA was extracted and sequenced to detect the presence of WT and/or mutant alleles. Three independently derived WT, three homozygous mutant Smchd1-KO clones, three homozygous Dux-knockout clones, three homozygous Smchd1/Dux double-knockout clones, and three homozygous mutant Tet1/Tet2/Tet3/Smchd1 quadruple-knockout clones were selected and used in this study.

For mammalian expression vectors, the Tet3FL, Tet3S, and Tet1 expression vectors were constructed as previously described (31). The pEF-Smchd1-FLAG expression vector was a gift from M. Blewitt. The pFastBac1-hTET2-CS construct was provided by R. Kohli (61). Fragments of TET3 and SMCHD1 were cloned into pEF-DEST51 expression vectors (Invitrogen, 430106). For co-IP of exogenously expressed full-length proteins, 293T cells were transfected by using a BioT transfection regent with plasmids expressing the appropriate FLAG- or V5-tagged proteins (5 g of each plasmid on a 10-cm dish). 293T cells were harvested at 48 hours after transfection, and nuclear lysates were purified by NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Fisher Scientific, 78835) according to the manufacturers instructions. The nuclear lysates were incubated with 2 g of the appropriate antibody for 2 hours and then incubated with 20 l of Dynabeads Protein G (Invitrogen, 00671375) overnight to collect the immune complexes. We washed the immune complexes with ice-cold wash buffer containing 10 mM tris-HCl (pH 7.4), 150 mM NaCl, 0.125% (v/v) NP-40, cOmplete (1 tablet/10 ml), and 2.5 mM EDTA. The samples were boiled in SDS-PAGE loading buffer, followed by SDS-PAGE, and Western blotting. For co-IP of protein domains, 293T cells were transfected with plasmids expressing the appropriate FLAG- or V5-tagged proteins (5 g of each plasmid on a 10-cm dish). 293T cells were harvested 48 hours after transfection and lysed in IP buffer [10 mM tris-HCl (pH 7.4), 150 mM NaCl, 0.125% NP-40, and 2.5 mM EDTA], supplemented with protease inhibitor cocktail (Roche).The cell lysate was centrifuged at 12,000g for 15 to 30 min at 4C, incubated with 2 g of the appropriate antibody for 2 hours, and then incubated with 20 l of Dynabeads Protein G (Invitrogen, 00671375) overnight. The beads were then washed six times with IP buffer [10 mM tris-HCl (pH 7.4), 150 mM NaCl, 0.125% NP-40, and 2.5 mM EDTA]. Last, the samples were boiled in SDS-PAGE sample loading buffer, followed by SDS-PAGE, and Western blotting with the indicated antibodies. For endogenous co-IP, preparation of nuclear extract, buffer preparation, and co-IP, we used the Nuclear Complex Co-IP Kit (Active Motif, 54001) according to the manufacturers instructions. We used 5 g of antibody for endogenous co-IP: SMCHD1 (Bethyl, A302-871A), TET1 antibody (GeneTex, GTX124207), TET2 antibody (Cell Signaling Technology, 92529), and TET3 antibody (31). After the IP reactions, we performed Western blotting with the indicated antibodies.

ES cells were washed twice with phosphate-buffered saline (PBS) and fixed in 4% paraformaldehyde in PBS for 15 min at room temperature. The fixed cells were permeabilized in 0.4% Triton X-100 in PBS at room temperature for 30 min, washed twice with PBS, and blocked for 30 min with 1% BSA in PBS. Cells were then incubated for 1 hour with anti-ZSCAN4 antibody (1:1000; Millipore, ab4340). After washing several times in 0.05% Tween 20 in PBS, the cells were incubated with Alexa Fluor 488 goat anti-rabbit (1:1000; Invitrogen, A27034) secondary antibody at room temperature for 1 hour and washed again three times. Then, we treated the cells with ProLong Gold anti-fade reagent with DAPI (4,6-diamidino-2-phenylindole) (Invitrogen, P36935) and acquired fluorescence images using a Nikon TE300 microscope with NIS-Elements AR 4.20.01.

FACS analysis was performed with a Beckman cell sorting system. Smchd1-KO mESCs or WT cells containing the 2C::tdTomato reporter (Addgene plasmid no. 40281; a gift from S. Pfaff) were subjected to FACS sorting with a MoFlo Astrios instrument (Beckman Coulter).

We seeded 1 105 293T cells into 24-well plates before transfection. Cells were transfected with the TET3S and SMCHD1 expression vectors, 47.5 ng of pGL3 luciferase reporter vector (methylated or unmethylated), and 2.5 ng of internal control Renilla luciferase reporter vector (pRL-CMV, Promega, Madison, WI). We harvested the cells 48 hours after transfection. All transfections were carried out at least in three independent experiments and in triplicate. Firefly and Renilla luciferase activities were assayed with the Dual-Luciferase assay kit (Promega) according to the manufacturers instructions. The firefly luciferase activities were normalized relative to Renilla activity.

BiFC assays were performed for determining the in vivo interaction between SMCHD1 and TET3. The assay is based on interactions between bait and prey proteins that bring together two nonfluorescent fragments of a fluorescent protein (GFP) and then form a functional chromophore. In this study, all recombinant expression vectors were constructed on empty backbones of HA-GFP1-10-pDEST-C and FLAG-GFP11-pDEST-C (Addgene plasmid nos. 118369 and 118367; gifts from M. Vartiainen). We used human embryonic kidney 293T cells, which were cotransfected with pEF-SMCHD1-HA-GFP1-10 and pEF-TET3-FLAG-GFP11 expression vectors using the BioT transfection regent. At 48 hours after transfection, the cells were analyzed by confocal microscopy (Zeiss LSM 880 microscope) and by Cell Cytometer Counter (Celigo) to identify the interactions. Cotransfection of pEF-OGT-HA-GFP1-10 and pEF-TET3-FLAG-GFP11 was used as a P.C. Cotransfection of pEF-SMCHD1-HA-GFP1-10 and pEF-ccdB-FLAG-GFP11 and cotransfection of pEF-ccdB-HA-GFP1-10 and pEF-TET3-FLAG-GFP11 were the N.C.s.

AlphaScreen (PerkinElmer) assays were performed for determining the in vitro interaction between biotin-SMCHD1 and His-tagged TET proteins following the manufacturers protocol. Briefly, 200 nM SMCHD1-biotin and 200 nM TET2FL-His or TET2-CD-His were incubated at room temperature for 1 hour. The protein sample was then incubated with streptavidin-coated donor beads (final concentration of 10 g/ml) and nickel-chelate acceptor beads (final concentration of 10 g/ml) in a total volume of 100 l of AlphaScreen buffer containing 50 mM MOPS (pH 7.4), 50 mM NaF, 50 mM 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate, and BSA (0.1 mg/ml) for 1 hour in the dark at room temperature. The photon counts were detected in 384-well plates by the EnVision Alpha reader (PerkinElmer).

Western blots and dots blot were performed as previously described (31), with minor modifications. For Western blots, we lysed the cells in buffer containing 50 mM tris-HCl (pH 7.4), 150 mM NaCl, 1% Triton X-100, 1 mM EDTA, and proteinase inhibitor cocktail (Roche, 11873580001) on ice for 60 min followed by centrifugation at 12,000g for 15 min at 4C. The lysates were separated on 4 to 15% SDS-polyacrylamide gels and transferred onto PVDF (polyvinylidene difluoride) membranes (Bio-Rad) by wet transfer at 4C. We incubated the membranes with blocking buffer (5% nonfat milk and 0.1% Tween 20 in PBS) for 1 hour at room temperature and then with the indicated primary antibody at 4C overnight. After washing with PBS-Tween (0.1%), the membranes were incubated with peroxidase-conjugated secondary antibodies for 1 hour at room temperature. Signals were detected using an ECL Prime detection reagent (GE Healthcare). Antibodies used for Western blots were as follows: anti-SMCHD1 (1:2500; Abcam, ab31865), anti-SMCHD1 (1:2500; Bethyl, A302-871A), anti-TET1 (1:1000; GeneTex, GTX124207), anti-TET2 (1:2000; Cell Signaling Technology, 92529) or anti-TET2 (1:1000; ProteinTech, 21207-1-AP), anti-DNMT1 (1:1000; Novus Biologicals, NB100-56519), anti-DNMT3A (1:1000; Novus Biologicals, NB120-13888), anti-DNMT3B (1:1000; Novus Biologicals, NB300-516), anti-tubulin (1:10,000; Abcam, ab7291), anti-ZSCAN4 (1:2500; Millipore, AB4340), HRP (horseradish peroxidase) goat anti-rabbit immunoglobulin G (IgG) (1:10,000; Active Motif, 15015), and HRP goat anti-mouse IgG (1:10,000; Active Motif, 15014). For dot blots, genomic DNA was purified with Quick-DNA Miniprep Plus kits (Zymo Research, D4070) followed by ribonuclease A treatment. The DNAs were then sonicated and purified using a QIAquick PCR purification kit (Qiagen, 28104). The purified DNAs were serially diluted and denatured in TE buffer at 98C for 10 min and then immediately chilled on ice for 10 min. The DNAs were then spotted onto a wetted GeneScreen Plus hybridization nylon membrane (PerkinElmer, NEF988001PK) with a Bio-Dot apparatus (96-well; Bio-Rad). The blotted membranes were ultraviolet cross-linked. After incubation with the blocking buffer (5% nonfat milk and 0.15% Tween 20 in PBS) for 2 hours at room temperature, the membranes were then incubated with anti-5hmC antibody (1:8000; Active Motif, 39769) or anti-5mC antibody (1:1000; Active Motif, 39649) for 1 hour at room temperature. After washing with PBS-Tween (0.15%), the membranes were incubated with peroxidase-conjugated secondary antibodies for 1 hour at room temperature. Signals were detected using an ECL Prime detection reagent (GE Healthcare).

DNA was purified with a Quick-DNA Miniprep Plus kit (Zymo Research, D4070). The bisulfite conversion was performed with the EZ DNA Methylation-Gold Kit (Zymo Research, D5005) according to the manufacturers instructions. PCR primer sequences for amplification of specific targets in bisulfite-treated DNA were 5TTTGTTAGGGATGAGGAGTT (forward) and 5AAACCTCTAATAAACCTCTTTA (reverse) for the Dux promoter. For sequence analysis, the PCR products obtained after bisulfite conversion were cloned into the Topo TA cloning vector (Thermo Fisher Scientific, 450030), and clones were sequenced. For TAB sequencing, 500 ng of genomic DNA was incubated with T4-glucosyltransferase (10 U/l) [New England Biolabs (NEB)], 2 mM uridine diphosphateglucose (NEB), and 10 CutSmart Buffer (NEB) at 37C overnight, and then DNA was purified using standard phenol chloroform extraction followed by ethanol precipitation. Next, we performed the TET oxidation reaction as follows: The purified DNA was incubated with 12.5 g of in-house purified TET2-CD protein, gelatin (1600 g/ml), TET oxidation buffer 1 [1.5 mM Fe(NH4)2(SO4)2], and TET oxidation buffer 2 [83 mM NaCl, 167 mM Hepes (pH 7.5), 4 mM ATP, 8.3 mM DTT, 3.3 mM -ketoglutarate, and 6.7 mM sodium ascorbate] at 37C for 2 hours. Then, we added 1 l of proteinase K (20 mg/ml) to the reaction, mixed well, and incubated at 50C for 10 min. We performed phenol/chloroform purification and ethanol precipitation and dissolved the purified DNA in TE buffer [10 mM tris-HCl (pH 8.0) and 0.1 mM EDTA]. Last, we performed the bisulfite conversion treatment of the purified DNA with the EZ DNA Methylation-Gold Kit (Zymo Research). For sequence analysis, the PCR products obtained after bisulfite conversion were cloned into the Topo TA cloning vector and clones were sequenced.

Total RNA was extracted from whole cells with a PureLink RNA mini kit (Ambion, 12183020), according to the manufacturers instructions. Total RNA integrity was verified with an Agilent 2100 Bioanalyzer (Agilent Technologies) and quantified with a NanoDrop 8000 instrument (Thermo Fisher Scientific). RNA-seq libraries were prepared from total RNA with the KAPA RNA HyperPrep kit with RiboErase (KAPA Biosystems). Library size distributions were validated on the Bioanalyzer (Agilent Technologies). Sequencing was performed with an Illumina NextSeq500 machine and 75-bp single-end reads were obtained. Library demultiplexing was performed following Illumina standards.

Trim Galore (version 0.4.0) was used to trim the 75-bp single-end reads. Reads were aligned to the mouse genome mm9 with STAR (version 2.5.1), and gene count was performed with STAR. Gene counts matrix was imported into R (version 3.5.1). Differential gene expression was determined with the Limma (version 3.38.2) statistical package19. Differential expression P values were adjusted for multiple testing correction using the Benjamini-Hochberg method in the stats package. Statistical significance for differentially expressed genes was fold change > 2 with q < 0.05. Heatmaps were generated with Pheatmap package. GSEA was performed with the GSEA preranked module of the Broad Institutes GenePattern algorithm (62). For the GSEA analysis, all data were compared with the 2c-like gene set of Macfarlan et al. (42). One thousand gene-list permutations were used to determine the FDR value and the classic scoring scheme, according to methods previously described (49). Repeat element analysis was done by calculating read counts falling completely within RepeatMasker-annotated repeat elements, and the density plot was generated with R.

For the WGBS library preparations, we used Swift, Accel-NGS Methyl-Seq DNA Library Kit (Swift Biosciences, 30024), and Zymos EZ DNA Methylation-Lightning kit (Zymo Research, D5030), according to the manufacturers instructions. Sequencing was performed with an Illumina HiSeq X with 150-bp paired-end read runs.

Paired-end whole-genome bisulfite reads were trimmed using TrimGalore!, version 0.5.0 (https://github.com/FelixKrueger/TrimGalore) with the following parameters to remove library preparation artifacts and low quality bases: --length 50, --clip_R1 10, --clip_R2 18, --three_prime_clip_R1 10, and --three_prime_clip_R2 10. Trimmed reads were aligned to the mm9 primary chromosomes using Bismark version 0.19.0 (63) and Bowtie2 version 2.3.3.1 (64) with the following parameters: -X 1000, --nucleotide_coverage, and --bowtie2. Duplicates were marked and removed using the deduplicate_bismark script provided with Bismark. CpG methylation values were extracted using the bismark_methylation_extractor script provided with Bismark and the following parameters: --no_overlap, --comprehensive, --merge_non_CpG, and --cytosine_report. We used DMRseq version 0.99.0 (41) to identify DMRs. Briefly, CpG loci with fewer than five reads were not considered for DMR calling, and a single CpG coefficient cutoff of 0.05 was used for candidate regions. Significant DMRs were identified using a q value < 0.05. Each CpG methylation value was averaged based on groups. t test showed a significant methylation difference between WT and KO (P < 2.2 1016). DMR-related genes were determined by defining a DMR within a genes proximity. DMRs were identified by bedtools (TSS 2K) and the Genomic Regions Enrichment Annotations Tool (GREAT) (65) and by long-range interaction between the DMRs and differentially expressed genes. We identified long-range interactions between the DMRs and differentially expressed genes by analyzing the Hi-C data in J1 ESC downloaded from GEO (Gene Expression Omnibus) dataset GSM862720 (SRR443885). Trim Galore (version 0.4.3) was used for adapter trimming for Hi-C data; HICCUP (version 0.5.9) was used for mapping and performing quality control. Significant interactions (default: P < 0.001 and z score > 1.0) were identified with HOMER, with a 40-kb resolution. Hi-C gene annotation involved identifying interactions with gene promoters, defined as 2 kb of a gene TSS (fig. S5G).

5hmC containing DNA was enriched by the EpiJET 5hmC Enrichment Kit (Thermo Fisher Scientific, K1491BID), according to the manufacturers instructions. The enriched DNA was then used for qPCR analysis of the Dux locus. qPCR reactions with target-specific primers included the forward (5GCTTTGCTACCAGGGAGGAG) and reverse (5GATCTTGAGCTGTGGGCCTG) primers for Dux region 1 and the forward (5CTAGCGACTTGCCCTCCTTC) and reverse (5GCTGATCAAGGAGGGGTTCC) primers for Dux region 2. PCR reactions were performed at 95C for 10 min followed by 50 cycles at 95C for 15 s, 57C for 30 s, and 72C for 30 s, using Power SYBR Green master mix (Applied Biosystems, 1809579) on a CFX96 real-time PCR cycler (Bio-Rad).

Total RNAs were isolated from cultured cells by using the PureLink RNA Mini Kit (Ambion). The SuperScriptIII reverse transcriptase (Invitrogen, 18080051) was used for reverse transcription of RNA, according to the manufacturers instructions. Real-time qPCR reactions with target-specific primers (available upon request) were performed at 50C for 2 min and 95C for 10 min followed by 50 cycles at 95C for 15 s and 60C for 1 min using TaqMan Gene Expression master mix (Applied Biosystems, 4369016) on a CFX96 real-time PCR cycler (Bio-Rad). The cDNA levels of target genes were analyzed using comparative Ct methods and normalized to internal standard, -actin.

ChIP was performed as previously described (31), with minor modifications. Briefly, cells were cross-linked with 1% formaldehyde (Thermo Fisher Scientific, 28908) in fixing buffer [50 mM Hepes (pH 7.5), 100 mM NaCl, 1 mM EDTA, and 0.5 mM EGTA] for 10 min at room temperature, and chromatin from lysed nuclei was sheared to 300- to 500-bp fragments using a Covaris E220 sonicator (Covaris; Woburn, MA). Chromatin fragments were incubated with 5 g of the appropriate antibody [SMCHD1 (Abcam, ab31865), TET1 (GeneTex, GTX125888), or IgG control (Santa Cruz Biotechnology; SC-2027)] overnight at 4C with rotation. For ChIP-qPCR, real-time qPCR was carried out with a CFX96 real-time PCR cycler (Bio-Rad). Each sample was analyzed in quadruplicate. Data were analyzed according to the 2(Ct of IP sample Ct of IgG sample) method and are presented as fold change of a percentage of input. PCR primer sequences are available upon request.

Acknowledgments: We thank G. Xu for providing Tet1/2/3 triple-knockout ES cells and H. Liu, P. Li, Z. Yuan, M. Du, K. Melcher, and S.-G. Jin for the advice and discussions. We thank the genomics, flow cytometry, high-throughput computing, and bioinformatics core facilities at the Van Andel Institute for the support. Funding: This work was supported by an Innovation Award from the Van Andel Institute. Author contributions: Z.H., J.Y., and G.P.P. designed and initiated the study and planned experiments; Z.H. performed interaction studies, genetic knockouts, and epigenomics and transcriptomics experiments; J.Y. and K.K. performed MS and analyzed the data; Z.H. analyzed the DNA methylation and gene expression data, with support from B.K.J.; W.C. provided experimental support; Z.H. and G.P.P. prepared the manuscript. All authors discussed the results and commented on the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. Genome-wide datasets generated in this study were deposited at the GEO database under the accession numbers GSE126468.

Go here to read the rest:
The chromosomal protein SMCHD1 regulates DNA methylation and the 2c-like state of embryonic stem cells by antagonizing TET proteins - Science Advances