Category Archives: Cell Biology

Fusion of maths and biology for coding human cells takes giant stride – Business Weekly

bit.bio, a Cambridge biomedical startup backed by Silicon Valley investors, has partnered with the London Institute for Mathematical Sciences, marking a milestone in the fusion of mathematics and biology for coding human cells.

The partnership, which aims to read out and reprogram all human cells like software, will challenge the conventional understanding of cell biology, mathematics and biological engineering.

The partnership heralds the fusion of previously separate industries such as software and synthetic biology to create a new industry in the industrial-scale production of human cells for biomedical testing and treatment.

Currently, cell therapies for diseases such as cancer are hampered by the lack of available human cells, while drugs tested on animals have a 97 per cent failure rate, partly due to differences between animal and human cells.

Synthetic biology, the redesign of organisms for new purposes, has been hindered by the difficulty in unlocking the fundamental laws governing cell identity.

This new partnership aims to unlock the operating system of life to facilitate the first mass-production of human cells for research and therapy purposes.

bit.bio, based at Babraham Research Campus, is backed by leading biotech investors who include Rick Klausner, the former US National Cancer Institute director.

It has already created the first large-scale, high-purity batches of neurons, muscle cells and oligodendrocytes and has developed a patented technique that could custom-build any human cell.

The company boasts a stellar scientific team including Dr Roger Pedersen, one of the pioneers of human stem cell biology and Dr Marius Wernig, a trailblazer in cell reprogramming and co-director of the Stanford Stem Cell Institute.

The London Institute for Mathematical Sciences is a private physics and maths research centre where scientists can commit themselves full-time to research. Its board includes a former chief scientific adviser to the Government and a former chief scientist at the Ministry of Defence.

The London Institute has previously been funded by DARPA to uncover fundamental laws in biology and has led pioneering work in similar fields from the geometry of genome space to the models of genetic regulatory networks.

Dr Mark Kotter, founder and CEO of bit.bio, said: This collaboration is incredibly exciting as we work on a paradigm shift in biology, moving it from an observational to a predictive science.

Over the past decade have learned that biology can be viewed as a software. Our collaboration with LIMS will help to decode the operating system of life.

This will unlock opportunities, including a new generation of cell therapies for tackling diseases such as cancer and dementia, accelerating drug development and could even help us combat pandemics of the future.

Dr Thomas Fink, founder and director of the London institute for Mathematical Sciences, added: Life is the final frontier of mathematics and the marriage of maths and biology will change the face of both disciplines.

Decoding cellular identity will require entirely new kinds of mathematics, as well as a deeper understanding of machine learning.

Living organisms exhibit extraordinary concision and elegance, the hallmarks off mathematical structure. The human genome amounts to just three gigabytes of data. But viruses, a mere seven kilobytes, can redirect it by calling up just the right subroutines in a similar way to how modular software works. Uncovering the operating system of life could enable us to engineer human cells as readily as we do software.

Read more:
Fusion of maths and biology for coding human cells takes giant stride - Business Weekly

VGLL4 promotes osteoblast differentiation by antagonizing TEADs-inhibited Runx2 transcription – Science Advances

INTRODUCTION

Cleidocranial dysplasia (CCD) is a hereditary disease characterized by incomplete closure of the fontanelle, abnormal clavicle, short stature, and skeletal dysplasia. It has been reported that there are multiple Runx2 mutations in human CCD syndrome (1, 2). Mature osteoblasts defect and bone mineralization disorders were observed in Runx2-deficient mice. The Runx2-heterozygous mice show similar phenotypes to the CCD syndrome (24). RUNX2 triggers mesenchymal stem cells (MSCs) to differentiate into osteoblasts (3, 5). According to the skeletal pathology studies in humans and mice, it is important to accurately regulate Runx2 activity during bone formation and bone remodeling (6, 7). However, the molecular regulation of Runx2 activity remains to be further studied.

The evolutionarily conserved Hippo pathway is essential for tissue growth, organ size control, and cancer development (811). Numerous evidences revealed the important roles of Hippo components in regulating bone development and bone remodeling. YAP, the essential downstream effector of Hippo pathway, regulates multiple steps of chondrocyte differentiation during skeletal development and bone repair (12). YAP also promotes osteogenesis and suppresses adipogenic differentiation by regulating -catenin signaling (13). VGLL4, a member of the Vestigial-like family, acts as a transcriptional repressor of YAP-TEADs in the Hippo pathway (14). Our previous work found that VGLL4 suppressed lung cancer and gastric cancer progression by directly competing with YAP to bind TEADs through its two TDU (Tondu) domains (9, 15). We also found that VGLL4 played a critical role in heart valve development by regulating heart valve remodeling, maturation, and homeostasis (16). Moreover, our team found that VGLL4 regulated muscle regeneration in YAP-dependent manner at the proliferation stage and YAP-independent manner at the differentiation stage (17). Our previous studies suggest that VGLL4 plays an important role to regulate cell differentiation in multiple organs. However, the function of VGLL4 in skeletal formation and bone remodeling is unknown.

Here, we reveal the function of VGLL4 in osteoblast differentiation and bone development. Our in vivo data show that global knockout of Vgll4 results in a wide variety of skeletal defects similar to Runx2 heterozygote mice. Our in vitro studies reveal that VGLL4 deficiency strongly inhibits osteoblast differentiation. We further demonstrate that TEADs can bind to RUNX2, thereby inhibiting the transcriptional activity of RUNX2 independent of YAP binding. VGLL4 could relieve the inhibitory function of TEADs by breaking its interaction with RUNX2. In addition, deletion of VGLL4 in MSCs shows similar skeletal defects with the global Vgll4-deficient mice. Further studies show that knocking down TEADs or overexpressing RUNX2 in VGLL4-deficient osteoblasts reverses the inhibition of osteoblast differentiation.

To study the function of VGLL4 in bone, we first measured -galactosidase activity in Vgll4LacZ/+ mice (16). -Galactosidase activity was enriched in trabecular bones, cortical bones, cranial suture, and calvaria cultures (fig. S1, A to C). Furthermore, in bone marrow MSCs (BMSCs), Vgll4LacZ/+ mice displayed -galactosidase activity in osteoblast-like cells (fig. S1D). During osteoblast differentiation in vitro, osteoblast marker genes such as alkaline phosphatase (Alp) and Sp7 transcription factor (Osterix) were increased and peaked at day 7. Vgll4 showed similar trend in this process at both mRNA and protein levels (Fig. 1A and fig. S1, E and F). To further clarify the important role of VGLL4 in bone development, we used a Vgll4Vgll4-eGFP/+ reporter mouse line in which VGLL4enhanced green fluorescent protein (eGFP) fusion protein expression is under the control of the endogenous VGLL4 promoter, and GFP staining reflects VGLL4 expression pattern in skeletal tissues (16). GFP staining was performed at embryonic day 18.5, week 1, week 2, and week 4 stages. The results indicated that the VGLL4 expression level was increased during bone development (fig. S1G). In addition, VGLL4 was enriched in trabecular bones, cortical bones, chondrocytes, cranial suture, and calvaria (fig. S1, G and K to M). We then observed the colocalization of VGLL4-eGFP with markers of MSCs (CD105), osteoblasts [osteocalcin (OCN)], and chondrocytes [collagen 2a1 (Col2a1)] in long bone and calvaria (fig. S1, H to M). Next, we analyzed VGLL4 expression pattern during osteoblast development in vivo (fig. S1N), which was similar to Alp and Osterix expression patterns in mouse BMSCs of different ages. Together, both in vivo and in vitro data suggest that VGLL4 may play roles in osteoblast differentiation and bone development.

(A) Immunoblotting showed the expression profile of VGLL4 during osteoblast differentiation in C57BL/6J mouse BMSCs. Samples were collected at 0, 1, 4, 7, and 10 days after differentiation. (B) Skeletons of WT and Vgll4/ mice at postnatal day 1 (P1) were double-stained by Alizarin red/Alcian blue (n = 5). Scale bar, 5 mm. (C) Quantification of body length in (B). (D) Skull preparations from control and Vgll4/ mouse newborns were double-stained with Alizarin red and Alcian blue at P1. -QCT images of skulls were taken from control and Vgll4/ mice at P4. Scale bar, 5 mm. (E) Quantification of skull defect area in (D). (F) Clavicle preparations from control and Vgll4/ mouse newborns were double-stained with Alizarin red and Alcian blue at P1 and quantification of clavicle length. Scale bar, 5 mm. (G) Alp staining and Alizarin red staining of calvarial cells from WT and Vgll4/ mice after cultured in osteogenic medium. Scale bar, 3 mm. (H) Relative mRNA levels were quantified by RT-PCR. (I) Hematoxylin and eosin (H&E) staining of femur from WT and Vgll4/ mice at embryonic day 16.5. Scale bar, 125 m. (J) In situ hybridization for Col11 immunostaining. Scale bar, 125 m. In (C), (E), (F), and (H), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001, ns, no significance; unpaired Students t test. Photo credit: Jinlong Suo, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai.

To investigate the potential function of VGLL4 in bone, we next analyzed the phenotype of Vgll4 knockout (Vgll4/) mice (16). The newborn Vgll4 knockout mice were significantly smaller and underweight compared with their control littermates (Fig. 1, B and C, and fig. S2, A and B). In particular, the membranous ossification of the skull was impaired in Vgll4/ newborns compared with the control littermates (Fig. 1, D and E). Furthermore, Vgll4 knockout mice developed a marked dwarfism phenotype with short legs and short clavicles (Fig. 1, C and F). To assess the role of VGLL4 in osteoblast differentiation, calvarial cells from Vgll4/ mice and wild-type (WT) mice were cultured in osteogenic medium. The activity of Alp in the Vgll4 deletion group was significantly reduced at the seventh day of differentiation (Fig. 1G, top) and was markedly weakened over a 14-day culture period as revealed by Alizarin red S staining (Fig. 1G, bottom). The declined osteogenesis in Vgll4 knockout cells was confirmed by the decreased expression of a series of osteogenic marker genes (Fig. 1H), including Alp, Osterix, and collagen type1 1 (Col11). In addition, in Vgll4/ mice, bone development was severely impaired with remarkable decrease in bone length and almost a complete loss of bone ossification (Fig. 1I). Consistently, immunohistochemical analysis of bone tissue sections from embryos at embryonic day 14.5 further confirmed the defects of bone formation and impaired osteoblast differentiation in Vgll4/ mice (Fig. 1J). Together, our study suggests that VGLL4 is likely to regulate MSC fate by enhancing osteoblast differentiation.

Given that the smaller size of mice is often caused by dysplasia, we also paid attention to the development of cartilage after Vgll4 deletion. As we expected, cartilage development was delayed in Vgll4-deficient mice determined by Safranin O (SO) staining (fig. S2C). Immunohistochemical analysis of collagen X (Col X) further confirmed the delay of cartilage development in Vgll4/ mice (fig. S2D). However, additional experiments would be required to determine the regulatory mechanism behind the observed chondrodysplasia. Although dwarfism was observed and trabecular bones were significantly reduced in the adult Vgll4/ mice, no significant cartilage disorder was observed by SO staining (fig. S2E). In adults, bone is undergoing continuous bone remodeling, which involves bone formation by osteoblasts and bone resorption by osteoclasts. We speculated that Vgll4 deletion might lead to decreased osteoclast activity. To distinguish this possibility, we performed histological analysis by tartrate-resistant acid phosphatase (TRAP) staining to detect osteoclast activity. The results showed that osteoclast activity was comparable between Vgll4/ mice and their control littermates (fig. S2F). Together, our results suggest that the phenotypes observed in Vgll4/ mice are mainly due to the defect of osteoblast activity.

To further explore the role of Vgll4 in the commitment of MSCs to the fate of osteoblasts, we generated Prx1-cre; Vgll4floxp/floxp mice (hereafter Vgll4prx1 mice) (fig. S3A). Prx1-Cre activity is mainly restricted to limbs and craniofacial mesenchyme cells (18, 19). Western blot analysis confirmed that VGLL4 was knocked out in BMSCs (fig. S3B). Vgll4prx1 mice survived normally after birth and had normal fertility. However, Vgll4prx1 mice exhibited marked dwarfism that was independent of sex (Fig. 2, A and B, and fig. S3C), which was similar to the phenotype of Vgll4/ mice. In particular, the membranous ossification of the skull and clavicle was also impaired in Vgll4prx1 mouse newborns compared with control littermates (Fig. 2, C to E). To assess the role of VGLL4 in osteoblast differentiation, BMSCs from Vgll4prx1 and Vgll4fl/fl mice were cultured in osteogenic medium. Markedly decreased ALP activity and mineralization were observed in Vgll4prx1 mice (Fig. 2, F and G). The declined osteogenesis in Vgll4 knockout osteoblasts was also proved by the decreased expression of a series of osteogenic marker genes, including Alp, Osterix, and Col1a1 (Fig. 2H). Normal Runx2 expression was detected in Vgll4prx1 mice (Fig. 2H). To further verify the role of VGLL4 in osteoblast differentiation, BMSCs from Vgll4fl/fl mice were infected with GFP and Cre recombinase (Cre) lentivirus and then cultured in osteogenic medium. Vgll4fl/fl BMSCs infected with Cre lentivirus showed markedly decreased ALP activity and mineralization (fig. S4A). Reduced VGLL4 expression by Cre lentivirus was confirmed by reverse transcription polymerase chain reaction (RT-PCR) (fig. S4B). The declined osteogenesis was also proved by the decreased expression of a series of osteogenic marker genes, including Alp, Osterix, and Col1a1 (fig. S4B).

(A) Skeletons of Vgll4fl/fl and Vgll4prx1 mice at P1 were double-stained by Alizarin red and Alcian blue. Scale bar, 5 mm. (B) Quantification of body length in (A) (n = 6). (C) Skull and clavicle preparation from Vgll4fl/fl and Vgll4prx1 mouse newborns were double-stained with Alizarin red and Alcian blue at P1. Scale bars, 5 mm. (D) Quantification of the defect area of skulls in (C) (n = 6). (E) Quantification of clavicle length in (C) (n = 6). (F) Alp staining and Alizarin red staining of BMSCs from Vgll4fl/fl and Vgll4prx1 mice after cultured in osteogenic medium. Scale bars, 3 mm. (G) Alp activity was measured by phosphatase substrate assay. (H) Relative mRNA levels were quantified by RT-PCR. (I) 3D -QCT images of trabecular bone (top) and cortical bone (bottom) of distal femurs. (J to N) -QCT analysis for trabecular bone volume per tissue volume (BV/TV, Tb) (J), trabecular number (Tb.N/mm) (K), trabecular thickness (Tb.Th/mm) (L), trabecular separation (Tb.Sp/mm) (M), and cortical bone thickness (Cor.Th/mm) (N). (O) Representative images of von Kossa staining of 12-week-old Vgll4fl/fl and Vgll4prx1 mice. Scale bar, 500 m. (P) Representative images of calcein and Alizarin red S labeling of proximal tibia. Scale bar, 50 m. (Q) Quantification of MAR. (R and S) ELISA analysis of serum PINP (ng ml1) and CTX-1 (ng ml1) from 10-week-old Vgll4fl/fl and Vgll4prx1 mice (n = 5). In (B), (D), (E), (G), (H), (J) to (N), and (Q) to (S), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, no significance; unpaired Students t test. Photo credit: Jinlong Suo, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai.

We next performed PCNA (proliferating cell nuclear antigen) staining and MTT assay to detect whether VGLL4 influences cell proliferation during bone development. No significant differences were found after VGLL4 deletion (fig. S5, A to C). We also did not detect significant changes of proliferation-related genes and YAP downstream genes (fig. S5, D and E). We next performed TUNEL (terminal deoxynucleotidyl transferasemediated deoxyuridine triphosphate nick end labeling) staining to detect whether VGLL4 influences cell apoptosis. In addition, no significant differences were found after VGLL4 deletion (fig. S5, F and G).

To further determine the function of VGLL4 in skeletal system, we did micro-quantitative computed tomography (-QCT) analysis to compare the changes in bone-related elements in the long bones of Vgll4prx1 mice and control littermates. We found that the 3-month-old Vgll4prx1 mice showed decreased bone mass per tissue volume (BV/TV) relative to age-matched control littermates (Fig. 2, I and J). Further analysis showed a reduction in trabecular number (Tb.N) of Vgll4prx1 mice compared to control mice (Fig. 2K), which was accompanied by a decrease in trabecular thickness (Tb.Th) and an increase in trabecular separation (Tb.Sp) compared to control mice (Fig. 2, L and M). Vgll4prx1 mice also showed decreased cortical bone thickness (Cor.Th) relative to the Vgll4fl/fl mice (Fig. 2N). The von Kossa staining showed reduced bone mineral deposition in 3-month-old Vgll4prx1 mice (Fig. 2O). The mineral apposition rate (MAR) was also decreased in Vgll4prx1 mice compared with control littermates by fluorescent double labeling of the mineralizing front (Fig. 2, P and Q). Consistent with the decreased bone mass in Vgll4prx1 mice, the enzyme-linked immunosorbent assay (ELISA) assay of N-terminal propeptide of type I procollagen (PINP), a marker of bone formation, revealed a reduced bone formation rate in Vgll4prx1 mice (Fig. 2R). However, the ELISA assay of C-terminal telopeptide of collagen type 1 (CTX-1), a marker of bone resorption, showed that the bone resorption rate of Vgll4prx1 mice did not change significantly (Fig. 2S). Collectively, Vgll4 conditional knockout mice mimicked the main phenotypes of the global Vgll4 knockout mice, further indicating that VGLL4 specifically regulates bone mass by promoting osteoblast differentiation.

To further determine whether the abnormal osteogenesis in Vgll4prx1 mice was caused by a primary defect in osteoblast development, we generated an osteoblast-specific Osx-cre; Vgll4floxp/floxp mice (hereafter Vgll4Osx mice) by crossing Vgll4fl/fl mice with Osx-Cre mice, a line in which Cre expression is primarily restricted to osteoblast precursors (fig. S6A) (6, 20). Vgll4Osx mice survived normally after birth and had normal fertility, but exhibited marked dwarfism in comparison with Osx-Cre mice (fig. S6, B and C), which was similar to the phenotypes of Vgll4/ and Vgll4prx1 mice. In addition, the membranous ossification of the skull and clavicle was also impaired in Vgll4Osx mice compared with control littermates (fig. S6C). -QCT analysis further confirmed the osteogenic phenotype of Vgll4Osx mice (fig. S6, D to J). Hence, the Vgll4Osx mice summarized the defects observed in the Vgll4prx1 mice, thus supporting the conclusion that VGLL4 is necessary for the differentiation and function of committed osteoblast precursors.

We next worked to figure out the mechanism how VGLL4 controls bone mass and osteoblast differentiation. The pygmy and cranial closure disorders in Vgll4/ mice were similar to that of Runx2-heterozygous mice. We therefore examined the potential interaction between VGLL4 and RUNX2. However, coimmunoprecipitation experiments did not show the interaction between VGLL4 and RUNX2 (Fig. 3A). Previous studies showed that VGLL4 could compete with YAP for binding to TEADs (9). The TEAD family contains four highly homologous proteins (8), which is involved in the regulation of myoblast differentiation and muscle regeneration (21). We determined whether the binding of VGLL4 with RUNX2 requires TEADs. Coimmunoprecipitation experiments showed that RUNX2 and TEAD14 had almost equivalent interactions (Fig. 3B). Next, we investigated whether TEADs control the transcriptional activity of Runx2. We used the 6xOSE2-luciferase reporter system that is specifically activated by RUNX2 to verify the role of TEADs (22). We performed dual-luciferase reporter assay with 6xOSE2-luciferase and Renilla in C3H10T1/2 cells, and the results showed that TEAD14 significantly inhibited the activation of 6xOSE2-luciferase induced by RUNX2 (Fig. 3C). Consistently, knockdown of TEADs by small interfering RNAs (siRNAs) markedly enhanced both basic and RUNX2-induced 6xOSE2-luciferase activity (fig. S8A). TEAD family is highly conserved, which consists of an N-terminal TEA domain and a C-terminal YAP-binding domain (YBD) (Fig. 3D) (23). Glutathione S-transferase (GST) pull-down assay revealed the direct interaction between RUNX2 and TEAD4 (Fig. 3E). Moreover, both TEA and YBD domains of TEAD4 could bind to RUNX2 (Fig. 3, F and G).

(A) Coimmunoprecipitation experiments of RUNX2 and VGLL4 in HEK-293T cells. The arrow indicated IgG heavy chain. (B) Coimmunoprecipitation experiments of RUNX2 and TEAD14 in HEK-293T cells. The arrow indicated IgG heavy chain. (C) 6xOSE2-luciferase activity was determined in C3H10T1/2 cells cotransfected with RUNX2 and TEAD14. Data were calculated from three independent replicates. (D) Schematic illustration of the domain organization for TEAD4, TEAD4-Nt, and TEAD4-Ct. (E) GST pull-down (PD) analysis between purified GST-RUNX2 and HIS-SUMO-TEAD4 proteins. (F) GST pull-down analysis between purified GST-RUNX2 and HIS-SUMO-TEAD4-TEA proteins. (G) Lysates from HEK-293T cells with Flag and Flag-RUNX2 expressions were incubated with recombinant GST-TEAD4-YBD protein. GST pull-down assay showed the binding between RUNX2 and TEAD4-YBD. (H) Cells isolated from WT mice were infected with TEAD lentivirus. Osteoblast differentiation was evaluated by Alp staining and Alizarin red staining after culture in osteoblast differentiation medium for 7 days (top) and 14 days (bottom). Data are representative of three independent experiments. Scale bars, 3 mm. (I) Alp activity quantification was measured by phosphatase substrate assay (n = 3). (J) Relative mRNA levels of Alp, Col11, and Osterix were quantified by RT-PCR. (K) Cells isolated from WT mice were infected with TEAD shRNA lentivirus. Osteoblast differentiation was evaluated by Alp staining and Alizarin red staining after culture in osteoblast differentiation medium for 7 days (top) and 14 days (bottom). Data are representative of three independent experiments. Scale bars, 3 mm. (L) Alp activity quantification was measured by phosphatase substrate assay (n = 3). (M) Relative mRNA levels of Runx2, Alp, Col11, and Osterix were quantified by RT-PCR. (N) Relative mRNA levels of Tead1-4 were quantified by RT-PCR. In (C), (I), (J), and (L) to (N), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, no significance; unpaired Students t test.

To determine whether overexpression of TEAD14 affects osteoblast differentiation, BMSCs from WT mice were infected with TEAD14 lentivirus and then cultured in osteogenic medium. The activities of ALP in TEAD14 overexpression groups were significantly reduced at the seventh day of differentiation [Fig. 3, H (top) and I] and were significantly weakened by Alizarin red S staining over a 14-day culture period (Fig. 3H, bottom). The declined osteogenesis in TEAD14 overexpression cells was confirmed again by the decreased expression of a series of osteogenic marker genes, including Alp, Col11, and Osterix (Fig. 3J). Next, we blocked the total activities of TEAD14 by short hairpin RNA (shRNA) lentiviral infection (Fig. 3N). The activity of Alp in TEAD14 knockdown group was significantly increased [Fig. 3, K (top) and L]. Over a 14-day culture period, osteogenic differentiation was significantly enhanced by Alizarin red S staining (Fig. 3K, bottom). The enhanced osteogenesis in TEAD14 knockdown cells was further confirmed by elevated expression of a series of osteogenic marker genes, including Alp, Col11, and Osterix (Fig. 3M). These results suggest that TEAD14 act as repressors of RUNX2 to inhibit osteoblast differentiation.

To investigate the mechanistic role of VGLL4 in inhibiting osteoblast differentiation, we then verified whether VGLL4 could affect the interaction between TEADs and RUNX2. We found that VGLL4 reduced the interaction between RUNX2 and TEADs (Fig. 4A). To further illustrate the relationship between RUNX2/TEADs/VGLL4, we checked the interaction between RUNX2 and TEADs in the BMSC of Vgll4fl/fl mice treated with GFP or Cre lentivirus. We found that the interaction between RUNX2 and TEADs was enhanced in Cre-treated cells (Fig. 4B). We noticed that there were conserved binding sites of RUNX2 (5-AACCAC-3) and TEAD (5-CATTCC-3) in the promoter regions of Alpi, Osx, and Col1a1, which are three target genes of RUNX2 (17, 24). We performed TEAD4 and RUNX2 chromatin immunoprecipitation (ChIP) assays in BMSCs. The results indicated that both TEAD4 and RUNX2 bound on Alp, Osx, and Col1a1 promoters (fig. S7, A to I). VGLL4 was a transcriptional cofactor, which could not bind DNA directly. We have demonstrated that VGLL4 promoted RUNX2 activity by competing for its binding to TEADs. Consistently, VGLL4 partially blocked TEADs-repressed transcriptional activity of RUNX2 (Fig. 4C). However, overexpression of VGLL4 in TEADs knockdown cells showed no marked change on RUNX2-induced 6xOSE2-luciferase activity compared with TEAD knockdown (fig. S8B). We then asked whether loss of VGLL4-induced disorders of osteoblast differentiation is related to TEADs. We knocked down TEADs by lentiviral infection in Vgll4-deficient BMSCs and then induced these cells for osteogenic differentiation. The differentiation disorders caused by VGLL4 deletion were restored after TEAD knockdown (Fig. 4, D to F). These data supported that VGLL4 released the inhibition of TEADs on RUNX2, thereby promoting osteoblast differentiation.

(A) Coimmunoprecipitation experiments of RUNX2, TEADs, and VGLL4 in HEK-293T cells. The arrow indicated IgG heavy chain. (B) Coimmunoprecipitation experiments of RUNX2 and TEADs in BMSCs cells of Vgll4fl/fl mice treated with GFP and Cre lentivirus. (C) 6xOSE2-luciferase activity was determined in C3H10T1/2 cells cotransfected with RUNX2, TEADs, and VGLL4. (D) Cells isolated from Vgll4fl/fl and Vgll4prx1 mice were infected with GFP and TEAD shRNA lentivirus. Osteoblast differentiation was evaluated by Alp staining and Alizarin red staining after culture in osteoblast differentiation medium for 7 days (top) and 14 days (bottom). Data are representative of three independent experiments. Scale bars, 3 mm. (E) Alp activity quantification was measured by phosphatase substrate assay (n = 3). (F) Relative mRNA levels of Vgll4, Runx2, Alp, Col11, and Osterix were quantified by RT-PCR. In (B), (D), and (E), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, no significance; unpaired Students t test.

YAP, the key transcription cofactor in the Hippo pathway, has been widely reported in regulating bone development and bone mass (12, 13). VGLL4, a previously identified YAP antagonist, directly competes with YAP for binding to TEADs (9). Therefore, we suspected that the inhibition of RUNX2 transcriptional activity caused by VGLL4 deletion might be dependent on YAP. To this end, we validated the role of YAP by 6xOSE2-luciferase reporter system. The data showed that YAP promoted RUNX2 activity in a dose-dependent manner (Fig. 5A). Moreover, TEAD4 significantly inhibited 6xOSE2-luciferase activity induced by YAP (Fig. 5B). TEAD4Y429H, a mutation that impairs the interaction between TEAD4 and YAP/TAZ (Fig. 5C) (25), did not promote 3xSd-luciferase activity induced by YAP (Fig. 5D). We found that both TEAD and TEAD4Y429H could interact with RUNX2 (Fig. 5E), and both TEAD4 and TEAD4Y429H could inhibit the activity of RUNX2 in a dose-dependent manner (Fig. 5, F and G). Restoring the expression of both TEAD4 and TEAD4Y429H could reverse the increased osteoblast differentiation in TEAD knockdown BMSCs (Fig. 5, H and I). Furthermore, overexpression of TEAD1 could further inhibit osteogenic differentiation of BMSCs after YAP knockdown (Fig. 5J). Together, these data suggest that the inhibition of RUNX2 activity by TEADs is independent of YAP binding.

(A) Effects of YAP on Runx2-activated 6xOSE2-luciferase activity in C3H10T1/2 cells. (B) 6xOSE2-luciferase activity was determined in C3H10T1/2 cells cotransfected with RUNX2, YAP, and TEAD4. (C) Schematic illustration of TEAD4 and TEAD4Y429H mutation. (D) 3xSd-luciferase activity was determined in HEK-293T cells cotransfected with YAP, TEAD4, and TEAD4Y429H. (E) Coimmunoprecipitation experiments of RUNX2, TEAD4, and TEAD4Y429H in HEK-293T cells. The arrow indicated IgG heavy chain. (F) Effects of TEAD4 on RUNX2-activated 6xOSE2-luciferase activity in C3H10T1/2 cells. (G) Effects of TEAD4Y429H on RUNX2-activated 6xOSE2-luciferase activity in C3H10T1/2 cells. (H) Cells isolated from WT mice were infected with GFP or TEAD shRNAs, TEAD4, or TEAD4Y429H lentivirus. Osteoblast differentiation was evaluated by Alp staining and Alizarin red staining after culture in osteoblast differentiation medium for 7 days (top) and 14 days (bottom). Data are representative of three independent experiments. Scale bars, 3 mm. (I) Alp activity quantification was measured by phosphatase substrate assay (n = 3). (J) Relative mRNA levels of Runx2, Alp, Col11, Osterix, Tead1, and Yap were quantified by RT-PCR. In (A), (B), (D), (F), (G), (I), and (J), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, no significance; unpaired Students t test.

We next examined how VGLL4 breaks the interaction between RUNX2 and TEADs. It has been reported that VGLL4 relies on its own two TDU domains to interact with TEADs (9), and VGLL4 HF4A mutation can disrupt the interaction between VGLL4 and TEADs (15). We hypothesized that VGLL4 competes with RUNX2 for TEAD1 binding depending on its TDU domain. On the basis of these previous studies, we performed coimmunoprecipitation experiments and found that VGLL4 HF4A abolished the interaction between VGLL4 and TEAD1 but did not affect the interaction between TEAD1 and RUNX2 (Fig. 6A). VGLL4 partially rescued the inhibition of RUNX2 transcriptional activity by TEAD1; however, VGLL4 HF4A lost this function (Fig. 6B). We then overexpressed TEAD1 by lentivirus infection in primary calvarial cells and found that the transcriptional level of Alp was significantly inhibited. This inhibition was released by overexpressing VGLL4 but not VGLL4 HF4A (Fig. 6C). To further verify the specific regulation of RUNX2 activity by VGLL4, we performed a coimmunoprecipitation experiment with low and high doses of VGLL4 and VGLL4 HF4A. The results showed that the TEAD1-RUNX2 interaction was gradually repressed along with an increasing dose of VGLL4 but not VGLL4 HF4A (Fig. 6D). Similarly, the inhibition of RUNX2 transcriptional activity by TEAD1 was gradually released with an increasing dose of VGLL4 but not VGLL4 HF4A (Fig. 6E). Super-TDU, a peptide mimicking VGLL4, could also reduce the interaction between purified RUNX2 and TEAD4 proteins (Fig. 6F). Thus, these findings suggest that VGLL4 TDU domain competes with RUNX2 for TEADs binding to release RUNX2 transcriptional activity.

(A) Coimmunoprecipitation experiments of RUNX2, TEAD1, VGLL4, and VGLL4 HF4A in HEK-293T cells. The arrow indicated IgG heavy chain. (B) 6xOSE2-luciferase activity was determined in C3H10T1/2 cells cotransfected with RUNX2, VGLL4, VGLL4 HF4A, and TEAD1 (n = 3). (C) RT-PCR analysis of Alp expression in calvarial cells. Cells isolated from WT mice were infected with GFP, TEAD1, VGLL4, or VGLL4 HF4A lentivirus. (D) Coimmunoprecipitation experiments of RUNX2, TEAD1, and an increasing amount of VGLL4 or VGLL4 HF4A in HEK-293T cells. The arrow indicated IgG heavy chain. (E) 6xOSE2-luciferase activity was determined in C3H10T1/2 cells cotransfected with RUNX2, TEAD1, and an increasing amount of VGLL4 or VGLL4 HF4A. (F) Competitive GST pull-down assay to detect the effect of VGLL4 Super-TDU on the interaction between RUNX2 and TEAD4. (G) Cells isolated from Vgll4fl/fl and Vgll4prx1 mice were infected with GFP and RUNX2 lentivirus. Osteoblast differentiation was evaluated by Alp staining and Alizarin red staining after culture in osteoblast differentiation medium for 7 days (top) and 14 days (bottom). Data are representative of three independent experiments. Scale bars, 3 mm. (H) Alp activity quantification was measured by phosphatase substrate assay (n = 3). (I) Relative mRNA levels of Vgll4, Runx2, Alp, Col11, and Osterix were quantified by RT-PCR. (J) Schematic model of VGLL4/TEADs/RUNX2 in regulating osteogenic differentiation. In (B), (C), (E), (H), and (I), data were presented as means SEM; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, no significance; unpaired Students t test.

Furthermore, we overexpressed RUNX2 by lentivirus infection in Vgll4 knockout BMSCs during osteogenic differentiation, and we found that RUNX2 could significantly restore the osteogenic differentiation disorder caused by Vgll4 deletion (Fig. 6, G to I). Together, these data suggest a genetic interaction between VGLL4/TEADs/RUNX2 and provide evidences that RUNX2 overexpression rescues osteogenic differentiation disorders caused by VGLL4 deletion.

Collectively, our study demonstrates the important roles of VGLL4 in osteoblast differentiation, bone development, and bone homeostasis. In the early stage of osteoblast differentiation, TEADs interact with RUNX2 to inhibit its transcriptional activity in a YAP bindingindependent manner. During differentiation progress, VGLL4 expression gradually increases to dissociate the interaction between TEADs and RUNX2, thereby releasing the inhibition of RUNX2 transcriptional activity by TEADs and promoting osteoblasts differentiation (Fig. 6J).

Accumulating evidences have suggested that the Hippo pathway plays key roles in regulating organ size and tissue homeostasis (8, 10). However, the transcription factors TEADs have not been reported in skeletal development and bone-related diseases. VGLL4 functions as a new tumor suppressor gene, which has been reported to negatively regulate the YAP-TEADs transcriptional complex. Our previous studies show that VGLL4 plays important roles in many tissue homeostasis and organ development, such as heart and muscle (16, 17). In this study, we provide evidences to show that VGLL4 can break TEADs-mediated transcriptional inhibition of RUNX2 to promote osteoblast differentiation and bone development independent of YAP binding.

Overall, our studies establish the Vgll4-specific knockout mouse model in the skeletal system. We show that VGLL4 deletion in MSCs leads to abnormal osteogenic differentiation with delayed skull closure and reduced bone mass. Our data also reveal that VGLL4 deletion leads to chondrodysplasia. Recent researches identified that chondrocytes have the ability to transdifferentiate into osteoblasts (2628), suggesting the possibility that loss of VGLL4 might reduce or delay the pool of chondrocytes that differentiate into osteoblasts. We identify that VGLL4 regulates the RUNX2-TEADs transcriptional complex to control osteoblast differentiation and bone development. TEADs can bind to RUNX2 and inhibit its transcriptional activity in a YAP bindingindependent manner. Recent studies pointed out that reciprocal stabilization of ABL and TAZ regulates osteoblastogenesis through transcription factor RUNX2 (29); however, we found that TEAD4-Y429H, a mutation at the binding site of TAZ and TEAD (25, 30, 31), can still significantly inhibit the activity of RUNX2. Therefore, we consider that the way TEAD regulates RUNX2 may not depend on TAZ regulation. Further research found that VGLL4, but not VGLL4 HF4A, can alleviate the inhibition by influencing the binding between RUNX2 and TEADs. It is possible that VGLL4 might influence the structure organization of the RUNX2-TEAD complex to some extent. Structural information may be required to answer this question and may provide more insights into the mechanism of VGLL4 in osteogenic differentiation.

Previous studies showed that mutations in RUNX2 cause CCD and Runx2+/ mice show a CCD-like phenotype. However, many patients with CCD do not have RUNX2 mutations. Our study may provide clues to the pathogenesis of these patients. A significant reduction of bone mass was observed in the adult mice, suggesting that VGLL4 and TEADs might be drug targets for treatment of cranial closure disorders and osteoporosis. In addition, further investigation of the clinical correlation of VGLL4 and cleidocranial dysplasia in a larger cohort will provide more accurate information for bone research. Our work also provides clues to researchers who are studying the roles of VGLL4 in tumors or other diseases. RUNX2 is highly expressed in breast and prostate cancer cells. RUNX2 contributes to tumor growth in bone and the accompanying osteolytic diseases (32). The regulation of RUNX2 transcriptional activity by TEADs and VGLL4 is likely to play essential roles in tumor, bone metastasis, and osteolytic diseases. Our work may provide clues to researchers who are studying the role of VGLL4 in bone tumors.

We demonstrate that TEADs are involved in regulating osteoblast differentiation by overexpressing and knocking down the TEAD family in vitro. However, the exact roles of TEADs in vivo need to be further confirmed by generation of TEAD1/2/3/4 conditional knockout mice. In the follow-up work, we will continue to study the mechanism of TEADs in skeletal development and bone diseases. Overall, although there are still some shortcomings, our work has greatly contributed to understand the TEADs regulation of RUNX2 activity.

Our work defines the role of VGLL4 in regulating osteoblast differentiation and bone development, and identifies that TEADs function as repressors of RUNX2 to inhibit osteoblast differentiation. We propose a model that VGLL4 dissociates the combination between TEADs and RUNX2. It is not clear whether VGLL4 is also involved in regulating other transcription factors or signaling pathways in the process of osteoblast differentiation and bone development. If that is the case, how to achieve cooperation will be another interesting issue worthy of further study.

Vgll4Lacz/+ mice, Vgll4 knockout (Vgll4/) mice, Vgll4Vgll4-eGFP/+ mice, and Vgll4 conditional knockout (Vgll4fl/fl) mice were generated as previously described (16, 17), and Vgll4fl/fl mice were crossed with the Prx1-Cre and Osx-Cre strain to generate Vgll4prx1 and Vgll4Osx mice. All mice analyzed were maintained on the C57BL/6 background. All mice were monitored in a specific pathogenfree environment and treated in strict accordance with protocols approved by the Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences.

The following antibodies were used: anti-Osterix antibody (1:1000; Santa Cruz Biotechnology, SC133871), anti-RUNX2 antibodies (1:1000; Santa Cruz Biotechnology, SC-390351 and SC-10758), anti-Flag antibody (1:5000; Sigma-Aldrich, F-3165), anti-HA (hemagglutinin) antibody (1:2000; Santa Cruz Biotechnology, SC-7392), anti-HA antibody (1:1000; Sangon Biotech, D110004), anti-MYC antibody (1:1000; ABclonal Technology, AE010), anti-PCNA antibody (1:1000; Santa Cruz Biotechnology, SC-56), rabbit immunoglobulin G (IgG) (Santa Cruz Biotechnology, SC-2027), mouse IgG (Sigma-Aldrich, I5381), anti-VGLL4 antibody (1:1000; ABclonal, A18248), anti-TEAD1 antibody (1:1000; ABclonal, A6768), anti-TEAD2 antibody (1:1000; ABclonal, A15594), anti-TEAD3 antibody (1:1000; ABclonal, A7454), anti-TEAD4 antibody (1:1000; Abcam, ab58310), and antipan-TEAD (1:1000; Cell Signaling Technology, 13295).

Cells were cultured at 37C in humidified incubators containing an atmosphere of 5% CO2. Human embryonic kidney (HEK)293T cells were maintained in Dulbeccos Modified Eagle Medium (DMEM) (Corning, Corning, NY) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Gibco) solution. C3H10T1/2 cells were maintained in -minimum essential medium (-MEM) (Corning, Corning, NY) supplemented with 10% FBS and 1% penicillin/streptomycin (Gibco) solution. To induce differentiation of BMSC into osteoblasts, cells were cultured in -MEM containing 10% FBS, l-ascorbic acid (50 g/ml), and -glycerophosphate (1080 mg/ml). The osteoblast differentiation level assay was performed following a previously published method (33). To quantitate Alp activity, cells incubated with Alamar Blue to calculate cell numbers and then incubated with phosphatase substrate (Sigma-Aldrich, St. Louis, MO) dissolved in 6.5 mM Na2CO3, 18.5 mM NaHCO3, and 2 mM MgCl2 after washing by phosphate-buffered saline (PBS). Alp activity was then read with a luminometer (Envision). Bone nodule formation was stained with Alizarin red S solution (1 mg/ml; pH 5.5) after 14 days of induction.

We collected femurs and tibias from mice and flushed out the bone marrow cells with 10% FBS in PBS. All nuclear cells were seeded (2 106 cells per dish) in 100-mm culture dishes (Corning) and incubated at 37C under 5% CO2 conditions. After 48 hours, nonadherent cells were washed by PBS and adherent cells were cultured in -MEM (Corning, Corning, NY) supplemented with 10% FBS and 1% penicillin/streptomycin (Gibco) solution for an additional 5 days. Mouse BMSCs in passage one were used in this study.

Total RNA was isolated from cells with TRIzol reagent (T9424, Sigma-Aldrich), and first-strand complementary DNA (cDNA) was synthesized from 0.5 g of total RNA using the PrimeScript RT Reagent Kit (PR037A, TaKaRa). The real-time RT-PCR was performed with the Bio-Rad CFX96 System. Gene expression analysis from RT-PCR was quantified relative to Hprt.

C3H10T1/2 cells were seeded overnight at 1 105 cells per well into a 12-well plate and transfected by PEI (polyethylenimine linear) with a luciferase reporter plasmid along with various expression constructs, as indicated. All wells were supplemented with control empty expression vector plasmids to keep the total amount of DNA constant. At 36 to 48 hours after transfection, the cells were harvested and subjected to dual-luciferase reporter assays according to the manufacturers protocol (Promega).

293T cells were seeded at 1 107 cells per 10-cm dish and cultured overnight. At 36 to 48 hours after transfection with PEI, cells were harvested and washed with cold PBS following experimental treatments. Then, cells were lysed with EBC buffer [50 mM tris (pH 7.5), 120 mM NaCl, and 0.5% NP-40] containing protease inhibitor cocktail (1:100; MedChem Express, HY-K0010). After ultrasonication, lysates were subjected to immunoprecipitation with anti-Flag antibodies (M2, Sigma-Aldrich) at 4C overnight, followed by washing in lysis buffer, SDSpolyacrylamide gel electrophoresis (PAGE), and immunoblotting with the indicated antibody.

RUNX2 and TEAD4-YBD were cloned into pGEX-4T-1-GST vector and expressed in Escherichia coli BL21 (DE3) cells. TEAD4 and TEAD4-TEA were cloned into HT-pET-28a-HIS-SUMO vector and expressed in E. coli BL21 (DE3) cells. The two TDU domains of VGLL4 were cloned into HT-pET-28a-MBP vector and expressed in E. coli BL21 (DE3) cells. VGLL4 Super-TDU was designed as previously described (15). GST, HIS-SUMO, and MBP-fused proteins were purified by affinity chromatography as previously described (17). The input and output samples were loaded to SDS-PAGE and detected by Western blotting.

CalceinAlizarin red S labeling measuring bone formation rate was performed as previously described (33).

Preparation of skeletal tissue and -QCT analysis were performed as previously described (34). The mouse femurs isolated from age- and sex-matched mice were skinned and fixed in 70% ethanol. Scanning was performed with the -QCT SkyScan 1176 System (Bruker Biospin). The mouse femurs were scanned at a 9-m resolution for quantitative analysis. Three-dimensional (3D) images were reconstructed using a fixed threshold.

ChIP experiments were carried out in BMSCs according to a standard protocol. The cell lysate was sonicated for 20 min (30 s on, 30 s off), and chromatin was divided into fragments ranging mainly from 200 to 500 base pairs in length. Immunoprecipitation was then performed using antibodies against TEAD4 (Abcam, ab58310), RUNX2 (Santa Cruz Biotechnology, SC-10758), and normal IgG. The DNA immunoprecipitated by the antibodies was detected by RT-PCR. The primers used were as follows: Alp-OSE2-ChIP-qPCR-F (5-GTCTCCTGCCTGTGTTTCCACAGTG-3), Alp-OSE2-ChIP-qPCR-R (5-GAAGACGCCTGCTCTGTGGACTAGAG-3), Alp-TBS-ChIP-qPCR-F (5-CCTTGCATGTAAATGGTGGACATGG-3), Alp-TBS-ChIP-qPCR-R (5-TATCATAGTCACTGAGCACTCTCTTGCG-3), Osx-OSE2-ChIP-qPCR-F (5-TTAACTGCCAAGCCATCGCTCAAG-3), Osx-OSE2-ChIP-qPCR-R (5-CCTCTATGTGTGTATGTGTGTTTACCAAACATC-3), Osx-TBS-ChIP-qPCR-F (5-ATGCCAAGAGATCCCTCATTAGGGAC-3), Osx-TBS-ChIP-qPCR-R (5-AGCTTGGTGAGCACAGCAAAGACAC-3), Col1a1-TBS/OSE2-Chip-qPCR-F (5-CTCAGCCTCAGAGCTGTTATTTATTAGAAAGG-3), and Col1a1-TBS/OSE2-Chip-qPCR-R (5-TTAATCTGATTAGAACCTATCAGCTAAGCAGATG-3). TBS indicated TEAD binding sites.

Mouse TEAD1, TEAD2, TEAD3, and TEAD4 siRNAs and the control siRNA were synthesized from Shanghai Gene Pharma Co. Ltd., Shanghai, China. siRNA oligonucleotides were transfected in C3H10T1/2 by Lipofectamine RNAiMAX (Invitrogen) following the manufacturers instructions. Two pairs of siRNAs were used to perform experiments.

Hematoxylin and eosin stain and immunohistochemistry were performed as previously described (7). Tissue sections were used for TRAP staining according to the standard protocol. Tissues were fixed in 4% paraformaldehyde for 48 hours and incubated in 15% DEPC (diethyl pyrocarbonate)EDTA (pH 7.8) for decalcification. Then, specimens were embedded in paraffin and sectioned at 7 m. Immunofluorescence was performed as previously described (33). Sections were blocked in PBS with 10% horse serum and 0.1% Triton for 1 hour and then stained overnight with anti-PCNA antibody (SC-56). Donkey anti-rabbit Alexa Fluor 488 (1:1000; Molecular Probes, A21206) was used as secondary antibodies. DAPI (4,6-diamidino-2-phenylindole) (Sigma-Aldrich, D8417) was used for counterstaining. Slides were mounted with anti-fluorescence mounting medium (Dako, S3023), and images were acquired with a Leica SP5 and SP8 confocal microscope. For embryonic mice, 5-mm tissue sections were used for immunohistochemistry staining, DIG-labeled in situ hybridization (Roche), and immunohistochemical staining (Dako).

TUNEL staining for apoptosis testing was performed as provided by Promega (G3250).

MTT assay for cell viability was performed as provided by Thermo Fisher Scientific.

We determined serum concentrations of PINP using the Mouse PINP EIA Kit (YX-160930M) according to the instructions provided. In addition, we determined serum concentrations of CTX-1 using the Mouse CTX-1 EIA Kit (YX-032033M) according to the instructions provided.

Tissue sections were used for SO staining according to the standard protocol. After paraffin sections were dewaxed into water, they were acidified with 1% acetic acid for 10 s and then fast green for 2 min, acidified with 1% acetic acid for 10 s, stained with SO for 3 min and 95% ethanol for 5 s, and dried and sealed with neutral glue.

Statistical analysis was performed by unpaired, two-tailed Students t test for comparison between two groups using GraphPad Prism Software. A P value of less than 0.05 was considered statistically significant.

Acknowledgments: We thank A. McMahon (Harvard University, Boston, MA) for providing the Prx1-Cre mouse line. We thank the cell biology core facility and the animal core facility of Shanghai Institute of Biochemistry and Cell Biology for assistance. Funding: This work was supported by the National Natural Science Foundation of China (nos. 81725010, 31625017, 81672119, and 31530043), National Key Research and Development Program of China (2017YFA0103601 and 2019YFA0802001), Strategic Priority Research Program of Chinese Academy of Sciences (XDB19000000), Shanghai Leading Talents Program, Science and Technology Commission of Shanghai Municipality (19ZR1466300), and Youth Innovation Promotion Association CAS (2018004). Author contributions: Z.W., L.Z., and W.Z. conceived and supervised the study. J.S. conceived and designed the study, performed the experiments, analyzed the data, and wrote the manuscript. X.F. made the constructs, performed the in vitro pull-down assay and ChIP experiments, analyzed the data, and revised the manuscript. L.Z. and Z.W. provided genetic strains of mice. J.S. and Z.W. bred and analyzed Vgll4/ mice. J.L. and J.W. cultured the cells and made the constructs. W.Z., L.Z., X.F., and Z.W. edited 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.

View original post here:
VGLL4 promotes osteoblast differentiation by antagonizing TEADs-inhibited Runx2 transcription - Science Advances

Gene editing shows promise as tool to fight neuro disorder in babies, UNC study finds – WRAL Tech Wire

CHAPEL HILL Babies born with a faulty maternal copy of the UBE3A gene will develop Angelman syndrome, a severe neurodevelopmental disorderwith no cure and limited treatments. Now, for the first time, scientists at the UNC School of Medicine show that gene editing and gene therapy techniques can be used to restore UBE3A in human neuron cultures and treat deficits in an animal model of Angelman syndrome.

This work, published inNatureand led by senior author Mark Zylka, PhD, Director of theUNC Neuroscience Centerand W.R. Kenan, Jr. Distinguished ProfessorofCell Biology and Physiology, lays important groundwork for a long-lasting treatment or cure for this debilitating disease, as well as a therapeutic path forward for other single-gene disorders.

Our study shows how multiple symptoms associated with Angelman syndrome could be treated with a CRISPR-Cas9 gene therapy, Zylka said.And we are now pursuing this with help of clinicians at UNC-Chapel Hill.

Left: UBE3A gene is off. Right: Using CRISPR, the gene is expressed and neurons fire (yellow). UNC images

Angelman syndrome iscaused by a deletion or mutation of the maternal copy of the gene that encodes the ubiquitin protein ligase E3A (UBE3A). The paternal copy ofUBE3Ais typically silenced in neurons, so the loss of maternalUBE3Aresults in a complete absence of the UBE3A enzyme in most areas of the brain. Thats crucial because the enzyme targets proteins for degradation, a process that maintains normal function of brain cells. When that process goes awry, the result is Angelman syndrome, a brain disorder with symptoms that include severeintellectual and developmental disabilities, seizures, and problems with speech, balance, movement, and sleep.

Turning on the paternal copy ofUBE3Ais an attractive therapeutic strategy because it could reverse the underlying molecular deficiency of the disease, Zylka said. However, the paternal gene is silenced by a long strand of RNA, produced in the antisense orientation toUBE3A,which blocks production of the enzyme from the paternal copy of the gene.

Members of the Zylka lab, including postdoctoral fellows Justin Wolter, PhD, and Giulia Fragola, PhD, set out to devise a way to use CRISPR-Cas9 to restore the UBE3A enzyme to normal levels by disrupting the antisense RNA. Preliminary data in cell cultures were promising, and Zylka received grants fromthe NIH, theAngelman Syndrome Foundation, and the Simons Foundation to test their findings in human neurons and in a mouse model of the disease.

In theNaturepaper, co-first authors Wolter and Hanqian Mao, PhD, a postdoc in the Zylka lab, and UNC colleagues describe using an adeno-associated virus (AAV) gene therapy to deliver the Cas9 protein throughout the brain of embryonic mice that model Angelman syndrome. Because UBE3A is essential for normal brain development, early treatment is crucial. The researchers found that embryonic and early postnatal treatment rescued physical and behavioral phenotypes that model core deficits found in Angelman syndrome patients. Remarkably, a single neonatal injection of AAV unsilenced paternalUbe3afor at least 17 months, and the data suggest this effect is likely to be permanent. The researchers also demonstrated that this approach was effective in human neurons in culture.

We were blown away when we got these results, Zylka said. No other treatments currently being pursued for Angelman syndrome last this long, nor do they treat as many symptoms. I am confident others will eventually recognize the advantages of detecting the mutation that causes Angelman syndrome prenatally and treating shortly thereafter.

Wolter added, The results of treating early were very promising. Since we learned we could reduce the severity of Angelman syndrome in mice, we are now focused on refining our approach in ways that will be suitable for use in humans.

While working to translate this research into the clinic, the Zylka lab will collaborate with researchers at the Carolina Institute for Developmental Disabilities (CIDD)to identify symptoms in babies that have the genetic mutation that causes Angelman syndrome.

Zylkas lab is working with CIDD researchers led by CIDD director Joseph Piven, MD, to use brain imaging and behavior observations to identify symptoms associated with Angelman syndrome in infants. Anecdotal reports suggest these infants have difficulty feeding and reduced muscle tone, but these and other early symptoms have not been rigorously characterized to date.

The idea is to use genetic tests to identify babies that are likely to develop Angelman syndrome, treat prenatally or around the time of birth, and then use these early symptoms as endpoints to evaluate efficacy in a clinical trial, Zylka said. Our data and that of other groups clearly indicate that prenatal treatment has the potential to prevent Angelman syndrome from fully developing.

As part of theNaturestudy, the researchers also found that the gene therapy vector blocked the antisense RNA by integrating into the genome at sites cut by CRISPR-Cas9. This so-called gene trap could be exploited to disrupt other long non-coding RNAs and genes.

Zylka added, We are incredibly excited to keep this work moving forward with the hope of helping children and families overcome this debilitating condition. Support from the NIH, the Simons Foundation, and the Angelman Syndrome Foundation was essential for moving this work forward.

Along with Zylka, Wolter, and Mao, co-authors of the Nature paper are Giulia Fragola, PhD, postdoc in the Zylka lab at the time of this research; Jeremy Simon, PhD, research associate professor; James Krantz, Zylka lab research associate; Hannah Bazick, Zylka lab graduate student; Baris Oztemiz, Zylka lab research technician; and Jason Stein, PhD, assistant professor of genetics and member of the UNC Neuroscience Center; all at UNC-Chapel Hill.

This research was funded by grants from the National Institutes of Health, the Simons Foundation, the Angelman Syndrome Foundation, the Eshelman Institute for Innovation, and the Pfizer-NCBiotech Distinguished Postdoctoral Fellowship in Gene Therapy.

(C) UNC-CH

Read more:
Gene editing shows promise as tool to fight neuro disorder in babies, UNC study finds - WRAL Tech Wire

Celebrities and Astronauts to "Show up for Science" at the NYSCF Gala & Science Fair – PRNewswire

NEW YORK, Oct. 23, 2020 /PRNewswire/ --Hosted by CNN Chief Medical Correspondent Sanjay Gupta, MD and featuring appearances from world-renowned cellist Yo-Yo Ma, former New York Mayor Michael Bloomberg, Whoopi Goldberg, Martha Stewart, actors Annaleigh Ashford, Lilli Cooper, Jesse Tyler Ferguson, Santino Fontana, Victor Garber, Jane Krakowski, Kelli O'Hara, Billy Porter, Seth Rogen, and John Slattery, among others, The New York Stem Cell Foundation Research Institute's first-ever virtual gala is streaming online on the evening of October 27th. Open to the public and free for all who would like to watch, guests can register at http://www.nyscf.org/gala.

Under the theme "Show Up for Science," the program will re-imagine the Science Fair, a signature element of NYSCF's traditional gala at which guests meet and talk with NYSCF Research Institute scientists, turning it into an online experience as special celebrity guests like Garber, Krakowski, andFerguson interview scientists and learn about the latest updates and breakthroughs in stem cell research. The Gala will bring the world of the NYSCF Research Institute laboratories to life, this year in a multi-platform digital experience produced by Broadway Director Scott Ellis with Music by Tree Adams and Andrew Schuyler.

The Gala also honors the three 2020 NYSCF Stem Cell Heroes: internationally renowned architect Frank Gehry; bioethicist and patient advocate Brooke Ellison, PhD, and award-winning architect and designer David Rockwell.

The 2020 NYSCF Research Institute Gala and Science Fair experience will also include NASA astronauts Serena Aun-Chancellor, MD, PhD, and Peggy Whitson, PhD, talking about stem cell research in space and about what this can mean for stem cell research. In Garber's segment, he will talk with NYSCF Senior Vice President of Research Scott Noggle, PhD, on how stem cells can help find a cure for diabetes, while Krakowski will discuss the basics of stem cells and how they are being used to study and fight COVID-19 with NYSCF Vice President, Automation Systems & Stem Cell Biology Daniel Paull, PhD.

The program will also include Ferguson speaking with NYSCF Principal Investigator Laura Andres-Martin, PhD, about NYSCF's new cutting-edge research on women's reproductive cancers, and NYSCF scientists Howard Kim, PhD, and Cecile Terrenoire, PhD will also share an update on NYSCF's macular degeneration stem cell therapy.

The 75-minute program will focus on education about science, using entertainment as a means to help viewers understand scientific concepts. It will also feature a short performance in honor of Frank Gehry by cellist Yo-Yo Ma, and a comedy skit by Fontana and Ashford.

Frank Gehry is perhaps the world's most celebrated living architect, known for his designs of landmark buildings including the Guggenheim Museum in Bilbao, Spain; Walt Disney Concert Hall in Los Angeles; and Fondation Louis Vuitton in Paris. The Los Angeles-based architect, whose buildings are known for the sweeping shapes and dramatic silhouettes, has won the Pritzker Prize, long considered the architecture profession's equivalent of the Nobel, as well as the Praemium Imperiale in Japan and the Gold Medal of the American Institute of Architects and the Gold Medal of the Royal Institute of British Architects. Frank has also dedicated himself to philanthropic work throughout his long career with numerous pro bono design projects in the arts, and he has had a longstanding engagement with medical research. NYSCF is honored to count Frank as a steadfast supporter of its mission to accelerate better treatments and cures for the most devastating diseases of our time through stem cell research.

David Rockwell is the founder and President of the Rockwell Group, an interdisciplinary and innovative architecture and design firm based in New York with offices in Los Angeles and Madrid, with work that ranges from restaurant and hotel design to cultural and educational institutions, theaters, and stage sets, to self-initiated pro bono projects supporting New York City during times of crisis. Ten years ago, David conceptualized the first Science Fair at a NYSCF Gala, and for many years he designed the environment that simulates the NYSCF Research Institute Laboratories and allows benefit guests to meet and mingle with NYSCF scientists. He has received numerous awards and recognition for his groundbreaking work, including a Tony Award in 2016 for set design, and NYSCF is honored to recognize his dedication, creativity, and talent for making cutting-edge science come alive each fall.

Brooke Ellison, PhD, is an Associate Professor at Stony Brook University, and the Director of Education and Ethics at the Stony Brook Stem Cell Facility. An expert in stem cell research policy and ethics, and longtime patient and disability advocate, Brooke has dedicated her professional career to changing the perception of life-saving science. Paralyzed from the neck down and dependent on a ventilator since age eleven following an accident, Brook has surmounted innumerable hurdles to achieve her goals, including serving on the Empire State Stem Cell Board, which designed New York State's stem cell policy. NYSCF is proud to recognize Brooke's tireless efforts to bring the promise of stem cell research to fruition, and for her advocacy for people in need of better solutions.

"Scientific research is more important than ever, particularly now in the midst of a global pandemic," stated NYSCF Research Institute CEO and Founder Susan L. Solomon. "I am thrilled that we are able to bring our research to life through this digital medium, and to honor our three incredible stem cell heroes. Private philanthropy is the fuel that drives our scientific success, and the Gala and Science Fair celebration highlight both the importance of our work and the promise of stem cell research around the world."

About The New York Stem Cell Foundation Research Institute

The New York Stem Cell Foundation (NYSCF) Research Institute is an independent non-profit organization accelerating cures and better treatments for patients through stem cell research. The NYSCF global community includes over 190 researchers at leading institutions worldwide, including the NYSCF Druckenmiller Fellows, the NYSCF Robertson Investigators, the NYSCF Robertson Stem Cell Prize Recipients, and NYSCF Research Institute scientists and engineers. The NYSCF Research Institute is an acknowledged world leader in stem cell research and in developing pioneering stem cell technologies, including the NYSCF Global Stem Cell Array and in manufacturing stem cells for scientists around the globe. NYSCF focuses on translational research in an accelerator model designed to overcome barriers that slow discovery and replace silos with collaboration. For more information, visitwww.nyscf.org.

David McKeon212-365-7440[emailprotected]

SOURCE The New York Stem Cell Foundation

Home

More:
Celebrities and Astronauts to "Show up for Science" at the NYSCF Gala & Science Fair - PRNewswire

Meet the Researcher: Sarah Tsuruo 21 (CLAS) – UConn Today

Sarah Tsuruo came to the University of Connecticut knowing she was interested in biology, citing the positive influence of her high school science teachers. At UConn, she has turned that interest into a productive undergraduate research career.

Tsuruo is a biological sciences major with a double minor in molecular and cell biology and womens, gender and sexuality studies (WGSS). Tsuruo believes her WGSS minor helps inform her understanding of the biology field.

I love talking about the different intersections that no one talks about in biology, Tsuruo says.

Tsuruo knew she wanted to do research as an undergraduate and reached out to professors whose work aligned with her interests, including professor Daniel Bolnick in the Department of Ecology and Evolutionary Biology (EEB).

When everything matched up, I was really excited to join his lab, Tsuruo says. So here I am three years later.

In Bolnicks lab, Tsuruo studies the interplay between the immune and endocrine systems, which led to a Summer Undergraduate Research Fund (SURF) award from UConns Office of Undergraduate Research this past summer. Tsuruo continues to explore how a fibrosis immune response affects sex hormones in stickleback fish.

As someone who wants to pursue a career in medicine, Tsuruo is interested in how these sex-specific immune differences can be translated to humans.

I think the evolutionary and ecology aspects of my EEB lab provides a more holistic picture of health and how that translates to humans, Tsuruo says.

Tsuruo credits Bolnick with helping her understand the work when she first started.

Hes a really incredible mentor who takes the time to break things down, Tsuruo says.

For Tsuruo, this patience and close mentoring is one of the things that prevented her from becoming discouraged and helped her learn about research.

In STEM fields, a lot of undergraduates may be unsupported by the higher ups in the field, so its great to have a mentor who walks you through it, Tsuruo says.

Tsuruo also worked at UConn Health as a clinical research intern with professor Ernst Reichenberger, where she researched quality-of-life measures for keloid patients who suffer from abnormal, tumor-like growths on otherwise healthy skin.

Having the ability to bring a new topic into the research field Im working in and bringing more intersectionality is really exciting, Tsuruo says.

Tsuruos ability to bridge fields and topics is also evident in her work in the community. Through a Change Grant, Tsuruo developed a trauma-informed STEM curriculum for children in a domestic violence shelter this past summer. These children often have different classroom needs from those who have not experienced that kind of trauma. She says she was inspired to take on this challenge thanks to her experiences as a WGSS minor and STEM mentor at UConns Community Outreach 4H Vernon.

We have to understand how to interact with these kids as appropriate for what theyve been through in the way thats going to help them be the best they can be, Tsuruo says.

The curriculum included a focus on fostering connections between children to form a support network.

Its really useful for these kids to be accepted as they are, she says.

Tsuruo is currently working on her honors thesis where she devised her own question and is performing this independent project in Bolnicks lab.

Tsuruo is looking at the interaction between sex hormones and the immune system. She hopes to discover what tradeoffs organisms make in this interaction in order to optimize survival.

While research can be a challenging puzzle at times, Tsuruo says the end results make the effort worth it.

Once you finally get it, its so rewarding to complete work that is significant and is leading to something, Tsuruo observes.

After graduating from UConn, Tsuruo plans to go to medical school to become a pediatrician or OB-GYN. She intends to continue to incorporate research into her career with a special focus on the social determinants of health.

I think research is very important to my career, Tsuruo says.

Tsuruo recommends undergraduates interested in joining a lab take the time to find one that aligns with their interests.

Its really important to give yourself the confidence you need to pick something that fits your goals, Tsuruo says.

One thing Tsuruo has learned from her experiences at UConn is that research calls for determination.

You need to be a quick thinker, resolve problems, and keep working on it, Tsuruo says.

Students interested in learning more about research opportunities at UConn can check out virtual events during the Month of Discovery.

Follow UConn Research on Twitter & LinkedIn.

Go here to see the original:
Meet the Researcher: Sarah Tsuruo 21 (CLAS) - UConn Today

New blood cancer treatment works by selectively interfering with cancer cell signalling – Science Codex

University of Alberta scientists have identified the mechanism of action behind a new type of precision cancer drug for blood cancers that is set for human trials, according to research published today in Nature Communications.

The research team led by Luc Berthiaume, cell biology professor in the Faculty of Medicine & Dentistry, spent four years working to understand how the compound PCLX-001 targets enzymes that perform myristoylation, a cellular process in which the fatty acid myristate modifies proteins so they can move to membranes and become part of the cell signalling system.

"The enzymes that transfer myristate onto proteins are overexpressed in some cancer cells, meaning there's more of those enzymes, so they have long been thought of as a logical target for cancer treatment," said Berthiaume, who is also chief scientific officer and co-founder of Pacylex Pharmaceuticals, the U of A spinoff company developing the drug.

"Until now no one has done a thorough analysis of this hypothesis," Berthiaume said. "We actually found that several types of cancer cells have fewer of these enzymes, making them seemingly easier to kill with our lead drug."

To demonstrate this, the researchers tested the drug against 300 different cancer cell types. They reported that blood cancer cells including lymphomas and leukemia, which have fewer of the enzymes, are extremely sensitive to the drug. It also killed other types of cancer cells when given at a higher concentration.

The team found that the drug stopped B-cell lymphoma tumour survival signals, killed B-cell tumour cells in both test-tube and animal experiments, and left non-cancerous cells unharmed, Berthiaume said.

Having completed the necessary biosafety studies, Pacylex plans to initiate Phase 1 trials of PCLX-001 in lymphoma, leukemia, breast and colon cancer patients at the Cross Cancer Institute in Edmonton, the B.C. Cancer Centre in Vancouver and Princess Margaret Cancer Centre in Toronto later this year, Berthiaume said.

"We think PCLX-001 is a compound with a large therapeutic window that can kill the cancer cells at a much lower concentration than what is needed to kill normal cells," he said. "That is the holy grail of cancer therapies.""Because of the highly selective nature of our drug, it's often referred to as a precision medicine, and we anticipate minimal side-effects," he said.

See the rest here:
New blood cancer treatment works by selectively interfering with cancer cell signalling - Science Codex

ONCURIOUS I-O Portfolio Directed at Boosting T Cell Influx and Activity in Solid Tumors Announcing First Preclinical Proof of Concept in Proprietary…

Leuven, Belgium - October 22, 2020-6.45 PM CET, ONCURIOUS NV, a Belgium-based biotech company focused on developing innovative immune-oncology treatments, today presents its strategy directed at boosting T cell migration, infiltration and activity into solid tumors, and announcesthat it has reached preclinical proof of concept and has entered the lead optimization phase with its proprietary CCR8 Treg program.

This important milestone follows Oncurious decision to focus its development activities on a pipeline of promising novel immune-oncology targets and drug discovery projects.

Oncurious scientists, in collaboration with world-class immuno-oncology experts in T cell and endothelial cell biology Prof. Dr. Gabriele Bergers (VIB-KU Leuven), Prof. Dr. Massimiliano Mazzone (VIB-KU Leuven) and Prof. Dr. Jo Van Ginderachter (VIB-VUB), and the drug discovery unit at VIB, are building a pipeline of proprietary investigational I-O therapies withdistinct mode of actions.

The team has discovered a potent and diverse panel of leads targeting human CCR8, has reached preclinical proof of concept and is entering the final lead optimization stages nearing preclinical candidate selection. Oncurious is now accelerating its efforts towards initiation of preclinical development of the therapeutic antibody program in early 2021.

CCR8 has been validated to be a tumor-infiltrating Treg-specific marker in solid tumors in both patients and animal models, making it the preferred target for therapeutic depletion of Tregs in cancer.

Oncurious CCR8 leads have been generated using an antibody technology platformthat has been validated and used for more than a decade to generate high quality binders against G-protein coupled receptors. Molecules discovered using this technology were tested in several preclinical tumor models, and showed that targeting CCR8, depleted Tregs specifically in the tumor microenvironment and resulted in strong anti-tumor responses in monotherapy as well as in combination with anti-PD1. The treatments led to the establishment of immunological memory.

Next to the anti-CCR8 program, Oncurious is focusing on 2 other programs aimed at boosting anti-tumor T cell influx and activity in immune excluded tumors. Exclusion of T cells is an immunosuppressive mechanism commonly used by cancers to evade the immune system and as such is an attractive target for new therapeutic modalities.

The company expects to make further announcements regarding its progress in the near future.

Patrik De Haes, M.D., Executive Chairman of ONCURIOUS NVcomments, I am excited about the future of Oncurious based on its portfolio of novel, differentiated I-O therapies. The companys approach is to generate novel drug candidates to treat immune excluded solid tumors that are not adequately addressed by immune checkpoint inhibitors, which are rapidly becoming the mainstay of cancer therapy. Today we are announcing significant progress in both hit and lead generation against novel I-O targets, including CCR8, which we believe could play a key role in the development of innovative immunotherapies to overcome solid tumor resistance. Working with leading scientists in the field, we aim to be at the forefront of a new wave of the I-O revolution and are confident that by delivering on our goals we can build Oncurious into a company of significant value in the years ahead. We look forward to updating you on our further progress.

Jrme Van Biervliet, Managing Director at VIB comments: We are very pleased to see Oncurious anti-CCR8 antibody reach this important step towards preclinical development, not least thanks to the model collaboration with VIB Discovery Sciences. Side-by-side with the Oncurious team, they are accelerating a promising pipeline of investigational immuno-oncology therapies.

- END

For further information please contact:

About ONCURIOUS

Oncurious is a Belgium-based biotech company focused on developing innovative oncology treatmentsderived froma series of promising new targets that are designed to enhance T cell activity, migration and influx into resistant tumour sites, boosting the patients own immune response against cancer. The company's lead immuno oncology asset is a Treg-depleting asset, targeting a protein specifically expressed on Tregs in the tumour microenvironment. The program is in lead optimization. The company's two other programs are focused on T cell migration and are in the discovery stages.

Immuno-oncology made a major step forward in the treatment of cancer with the advent of immune checkpoint inhibitors, which are expected to represent a market opportunity of $50bn by 2025 (GlobalData 2020). Despite this clinical and commercial success, there remains an important need to develop new I-O approaches that improve efficacy, overcome resistance, and that can synergize with checkpoint inhibitors so that more patients with solid tumors can benefit from these life-saving therapies.

Oncurious' early stage pipeline of novel immune-oncology programs offer the potential to overcome tumor resistance mechanisms, that current immune checkpoint inhibitors cannot address, and thereby significantly enhance the responses to immunotherapy across multiple tumor types.

More information: http://www.oncurious.com

About VIBVIB is an excellence-based entrepreneurial research institute in life sciences located in Flanders, Belgium. VIBs basic research leads to new and innovative insights into normal and pathological life processes. It unites the expertise of all its collaborators and research groups in a single institute, firmly based on its close partnership with 5 Flemish universities (Ghent University, KU Leuven, University of Antwerp, Vrije Universiteit Brussel and Hasselt University) and supported by a solid funding program from the Flemish government.

VIB has an excellent track record on translating basic scientific results into pharmaceutical, agricultural and industrial applications. Since its foundation in 1996, VIB has created 25 start-up companies, now employing over 900 people. The link between basic research and valorisation has made VIB a catalyst for the ever-growing biotech hotspot in Flanders. In recent years, numerous biotech companies both large and small have settled down in the region, thanks to top-notch infrastructure set up and provided by VIB and the ready availability of new scientific talent from the VIB labs.

More information: http://www.vib.be

Original post:
ONCURIOUS I-O Portfolio Directed at Boosting T Cell Influx and Activity in Solid Tumors Announcing First Preclinical Proof of Concept in Proprietary...

USC biological imaging innovator elected to National Academy of Medicine > News > USC Dornsife – USC Dornsife College of Letters, Arts and…

Provost Professor Scott Fraser is recognized for developing technology that provides unprecedented views of live organisms, from embryonic development to old age. [3 min read]

Provost Professor Scott Fraser, a recognized innovator whose inventions have found wide use in both scientific and clinical settings, is an elected member of the National Academy of Medicine. (Photo: No Montes.)

Scott Fraser, Provost Professor of Biological Sciences, Biomedical Engineering, Physiology and Biophysics, Stem Cell Biology and Regenerative Medicine, Pediatrics, Radiology and Ophthalmology, has been elected to the National Academy of Medicine.

Fraser, who holds joint appointments at the USC Dornsife College of Letters, Arts and Sciences and USC Viterbi School of Engineering as well as the Elizabeth Garrett Chair in Convergent Bioscience, is one of just 90 researchers chosen from among the worlds leading scientists to become members of the academy.

Professor Scott E. Fraser is a brilliant biophysicist and innovator, said USC Provost Charles F. Zukoski. He is being recognized for groundbreaking advancements in biology and medicine. His research, which centers on imaging and molecular analyses of intact biological systems, serves as inspiration for future generations of engineers, scientists and medical professionals.

Among the reasons for his election, the academy noted Frasers work integrating biophysics, quantitative biology, and molecular imaging to enable unprecedented views of normal function and disease in live organisms, from embryonic development to old age.

Ive always been fascinated by interdisciplinary teams that can bring new insights into old problems by combining the insights from science, engineering and medicine, Fraser said.

Applying tricks from other fields

Fraser, who earned his bachelors degree in physics and his Ph.D. in biophysics, says he gravitated toward research in biology because there are so many open questions, and so many things that have been thought to be impossible to answer but tricks from other fields make the impossible possible, if the team is willing to tackle it together.

Frasers research delves into early development, organogenesis (the process by which internal organs emerge and develop) and medical diagnostics. His work has spawned several start-up companies and has been used in a number of instruments and FDA-approved diagnostics.

We keep our eyes open to translation of the work in the lab to industrial and clinical utility, he said, adding that USCs Alfred E. Mann Institute for Biomedical Engineering and USC Viterbis National Science Foundation-funded Innovation Corps node have both played key roles and offered important instruction on how to best bring their work to potential customers.

In the last year, our IP (intellectual property) has been licensed by a half-dozen different companies, he said. So, we know the work can lead to new instruments, new diagnostics and new techniques.

Fraser said his team works diligently to ensure collaborators in scientific and clinical fields also benefit from their efforts.

We have built the Translational Imaging Center on the University Park campus and the Translational Biomedical Imaging Center at Childrens Hospital Los Angeles to help support users with interests in fields ranging from regenerative medicine to cancer and diabetes. This is already empowering them to make new insights into their research challenges.

What we hope to do is to make it possible for researchers and clinicians to have aha moments, when they can see things for the first time.

A career highlighted by innovation

After earning his Ph.D. in 1979, Fraser joined the faculty at the University of California, Irvine, where he rose through the ranks to become chair of the Department of Physiology and Biophysics. In 1990, he moved to Caltech to serve as the Anna L. Rosen Professor of Biology and the director of the Biological Imaging Center. There, he served as the founding director of both the Caltech Brain Imaging Center and the Rosen Center for Biological Engineering and helped found the Kavli Nanoscience Institute.

In Fall 2012, Fraser moved to USC as Provost Professor at USC Dornsife and USC Viterbi, with formal links to Childrens Hospital Los Angeles and Keck School of Medicine of USC. He serves as the director of science initiatives for USC as well as co-director of the Bridge Institute at the USC Michelson Center for Convergent Bioscience.

A prolific author and inventor, Fraser has more than 240 peer-reviewed articles and more than 75 issued patents to his credit. He is the recipient of numerous honors and has been elected to the National Academy of Inventors, the American Institute for Medical and Biological Engineering, the American Association for the Advancement of Science, the American Academy of Arts and Sciences and the European Academy of Science.

About the National Academy of Medicine

The National Academy of Medicine, established in 1970 as the Institute of Medicine, is an independent organization of professionals from diverse fields including health and medicine, and the natural, social and behavioral sciences. Election to the academy recognizes individuals who have demonstrated outstanding professional achievement and commitment to service.

Read the rest here:
USC biological imaging innovator elected to National Academy of Medicine > News > USC Dornsife - USC Dornsife College of Letters, Arts and...

Homes Become Research Labs for Students Studying Opioid Addiction and Pain – Rutgers-Camden NewsNow

Nathan Fried and Ed Waddell assemble lab in a box supplies for students to conduct research at home.

By Jeanne Leong

According to the Centers for Disease Control and Prevention (CDC), opioid overdoses killed more than 46,000 people in 2018. Even in the midst of the global COVID-19 pandemic, opioid addiction remains a serious public health issue throughout the United States.

Students in a Rutgers UniversityCamden biology class are working to better understand the neuroscience related to this crisis, thanks to an innovative program that allows them to conduct research from home.

People are struggling with addiction and the loss of loved ones, says Nathan Fried, a Rutgers UniversityCamden assistant teaching professor of biology. It has touched personally each and every one of us.

Fried, a neuroscientist who researches chronic pain and how humans perceive pain, is leading a remote learning course for undergraduates this fall to study the neuroscience of pain and addiction to opioids. From basic cell function to how a neuron communicates from one location to another to deliver pain signals, RutgersCamden students in the Neuroscience of the Opioid Epidemic course are learning how addiction works within the brain.

The Neuroscience of the Opioid Epidemic class meets on Zoom for lectures and discussions about their research

The lab component of the course allows students to conduct research in their homes. Each of the 18 students mostly first-year students conducting research for the first time have received a lab in a box to set up a home laboratory to study pain and addiction in Drosophila melanogaster, the common fruit fly. The supplies, including fruit flies, a microscope, pipettes, and other tools, allow students to perform experiments as they would working in an on-campus lab.

Once they have learned a little bit about cell biology, neuroscience, chronic pain, and these mechanisms of addiction, they can start putting together this larger picture of all of the foundational things that are necessary to understand the opioid epidemic from a public health perspective, explains the RutgersCamden scientist.

Edward Waddell delivering lab in a box to students

Fried taught a similar in-person class before the pandemic, but now he and Edward Waddell, a University of Pennsylvania PennPORT IRACDA post-doctoral fellow who is teaching the course with Fried, designed the remote learning program to engage students through the challenges of learning virtually during the pandemic.

In this virtual environment, planning is essential, Fried said. I think its the same thing for students. When they were in the physical environment, they could just come in to class and see what we are doing today. So many people prepare on the fly, but in this virtual environment, we all have to plan extensively to be able to overcome the hurdles to make this a worthwhile experience.

I am grateful for the opportunity to do research as an undergraduate, says Bradley Mahler, a first-year history major from Laurel Springs. I am interested in how the seminar will address one of the issues that our country continues to deal with.

Bradley Mahler

Remote learning has been challenging for students and faculty, but Fried says he is surprised to see what he calls this wonderful energy flow happening in the class discussions that he usually experiences in an in-person class.

Students are going back and forth, they are highly engaged, and you see the gears moving in their heads, Fried explains. Students are still having those ah ha! moments, life-changing moments that change perspectives. Its gratifying to know that RutgersCamden can provide students with those crucial moments in this virtual environment.

Ashley Smigocki

RutgersCamden junior Ashley Smigocki, who plans a career in health care working with underserved populations, is interested in learning more about addictions.

I believe that the experience of scientific research and interactions with others in a scientific context will give me skills that I will apply for the rest of my academic journey, says Smigocki, a psychology major from Mount Laurel.

During the global pandemic, all national and international research conferences are now virtual, offering a unique opportunity for students in this Rutgers UniversityCamden class to attend a professional biology conference. In December, RutgersCamden students will participate in the conference of the American Society for Cell Biology and the European Molecular Biology Organization to hear from professionals in the discipline. Students will critique selected scientific talks from the conference for the final exam in Frieds course.

Read the original:
Homes Become Research Labs for Students Studying Opioid Addiction and Pain - Rutgers-Camden NewsNow

UConn faculty reflect on surveillance testing efforts, look to the future – UConn Daily Campus

Rachel ONeill, professor of molecular and cell biology, Kendra Maas, facility scientist at the University of Connecticuts MARS and Suzanne Onorato, executive director of student health and wellness, touched on the success of UConns fall testing efforts and discussed their hopes and plans for the spring.

ONeill began by providing a brief description of why the saliva testing effort is valuable. In essence, she said the pooled testing efforts were done to more efficiently utilize resources while working with UConns low positivity rate.

The overarching goal of this whole collective program is to reduce costs significantly and save critical, clinical-level tests for symptomatic or high-positivity rate areas, ONeill said. Right now, were at a really low positivity rate, so its an ideal approach to take.

Maas further emphasized UConns successful numbers so far. She said this reflects students general willingness to follow CDC guidelines and regulations.

I can say that, so far, the surveillance screening that we have done has been remarkably low positives, and thats been reflected by follow-up point of care tests and that percentage that were seeing at UConn, which is lower than the state, so we can say that students are doing a good job of following those CDC guidelines, Maas said.

In addition to the group testing being done, Onorato wanted to draw attention to Dr. Maass work with wastewater sampling. Onorato said they hope to expand this test to get even more accurate readings of where the COVID-19 increase may have originated.

When a particular wastewater site begins to have an increase in the presence of COVID, we then deploy pooled sampling to screen those students who reside in that area, Onorato said. This then allows for us to locate the positive cases early on and allows for rapid containment of the spread of COVID. We would like to continue to both create more local sources for wastewater testing in order to get even closer to the source with our pooled sampling.

ONeill said one of the major challenges they have faced so far has been clearly communicating everything to students. She said it is their responsibility to convey information to students well.

Weve actually done collaborations with amazing teams on campus who are going to help us translate this information and make it publicly available because I think it is our responsibility to translate that information and allow people to know this is the level when you should be worried, and this is more informational and we need to be keeping track, ONeill said.

ONeill also said it was important to note the expanse of the program and the amount of collaboration it has required. In particular, she noted the work done by UConn facilities to help Dr. Maas install her wastewater monitoring systems.

This project has been collaborative and huge. Its involved many, many groups on campus who have been incredible at stepping up to try to protect the safety of the students, ONeill said. Facilities have been right there every step of the way helping Kendra put these wastewater monitoring systems in place. Its just been absolutely incredible to watch.

Maas wanted to draw attention to the work being done by the student body as well. She said she has several UConn students, both graduate and undergraduate, assisting with her work.

All of the technicians who are doing this are students, Maas said. I have three masters grad students who do most of the lab work, and then I have a couple of undergrads who are helping collect samples and distribute tubing. This is all students.

Onorato spoke about future testing plans and the potential expansion of surveillance testing. She said this would likely be done in addition to educational efforts that showed proper strategies for the UConn student body regarding COVID-19.

ISG is also interested in expanding the surveillance testing strategy across campus in order to meet an increasing desire for expanded testing as well as promoting education to the broader student community regarding UConns COV2 strategy (through a new partnership with the newly establishedDxGroupat UConn), Onorato said. We are also considering a more routine pooled sampling approach for our residential student population.

Original post:
UConn faculty reflect on surveillance testing efforts, look to the future - UConn Daily Campus