Search Immortality Topics:

Page 7«..6789..2030..»


Category Archives: Genetic Medicine

Therapeutic Effects of Tretinoin | JIR – Dove Medical Press

Introduction

Sepsis is a lethal critical disease, identified as a main health concern across the world. In the past 10 years, hospital mortalities from sepsis and septic shock every year registered 17% and 26%, separately, causing about 8 million deaths each year.1,2 Cardiac function disorder induced by sepsis, often referred to as SIC, is commonly seen and has long been an intriguing topic.3 As a pathophysiological syndrome caused by infection rather than a specific disease, the specific identification of targeted SIC is essential for minimizing the mortality and morbidity in this regard. Despite the fact that the indications for supervising and healing SIC are clinical and directed toward restoring tissue perfusion, a deeper comprehension of the important gene signatures and underlying pathogenesis of SIC can assist in optimizing treatments and ameliorating clinical results.47

Gram-negative bacterial endotoxin (lipopolysaccharide, LPS) serves as a key sepsis mediator for septicemia-associated multiple organ dysfunction or mortality.8 After undergoing LPS insult, both macrophages and cardiomyocytes can release substantial inflammatory mediators, such as MCP-1, GM-CSF, IL-1, IL-1, IL-6, IL-7, IL-8, and IL-12.9,10 These inflammatory cytokines could give rise to the imbalance of calcium homeostasis,11 disturbance of energy metabolism,12 impairment of adrenergic signaling, and excess production of nitric oxide,13 all of which facilitate decreased contractility, diastolic dysfunction, impaired ejection fraction and reduced cardiac index. Cytokines, especially IL1, IL6, and TNF, are major contributors in the initiation of SIC.14

RA is a powerful derivant of vitamin A, and is pivotal for body developmental process and organ genesis via regulating cellular proliferative and differentiative activities. Substantial researches on animal models and clinic tests have verified its capability of preventing infections and enhance immunosystems.15,16 Austenaa et al demonstrated that RA inhibited LPS-triggered stimulation in mice and mankind monoblasts.17 In addition, Martire-Greco D et al displayed that all-trans retinoic acid (ATRA), which could improve functional immunoresponses in LPS-exposed mice, could serve as a novel underlying method for the healing of the immune suppressive status of sepsis.18 Considering the increasing evidence that RA can improve immune function and reduce inflammation, we speculated that RA might exert a beneficial effect on LPS-induced heart function disorder. The present research aimed to determine the roles of RA in LPS-induced cardiac dysfunction and explore its underlying mechanisms.

Currently, the network pharmacological approach can be employed to forecast the correlation between targets and diseases.19,20 The network pharmacological approach was deemed as a new method of medicine design.21,22 Network pharmacology methods are developing rapidly and have been leveraged to find new treatment methods, ameliorating the approved drugs and expanding the application scenarios of clinical medicines. They can also be utilized to effectively search for undeveloped targets for compounds or natural products.23 The purpose of network construction was to reveal the interaction between bioactive compounds and target proteins as well as the interaction between various target proteins. We identified and verified key nodes through network analysis and verification.21 Systematic or network pharmacology combined with multiomics analysis showed unique advantages in predicting and explaining the pharmacological principles of drugs and mechanisms of action in treating various diseases.24,25 For that reason, herein, network pharmacological approach was employed to discover treatment targets and associated signaling pathways of RA against SIC.

The primary aims of our research were 1) to select the underlying targets of Retinoic acid and DEGs in SIC heart tissues; 2) to study the potential causal links of Retinoic acid against SIC via bioinformatic analysis; 3) to confirm the anti-inflammation, antioxidant levels and potential signaling pathways of Retinoic acid in LPS-induced cardiac dysfunction. Our research might offer a novel treatment method for improving sepsis-induced cardiomyopathy.

Male C57BL/6 mice (810 weeks) were kept at the Experiment Animal Center of Wenzhou Medical University. Those animals were kept at 231 with a 12-hour light/dark period in a specially disinfected environment with free access to bacteria-free water and food. Every animal assay, with a minimum sacrificed mice according to our design, was accepted by the Animal Assay Ethical Board of our university (ID: WYYY-AEC-2021-301). All experiment procedures were completed blindly, such as the animal models and following assays. The animals were stochastically separated into these groups: (1) Saline (i.p., n= 6), (2) LPS(10mg/kg i.p., n= 6), (3) LPS(10mg/kg) plus RA(1mg/kg, i.p., n= 6), (4) LPS(10mg/kg) plus RA(3mg/kg, i.p., n= 6), (5) LPS(10mg/kg) plus Dexamethasone (DEX)(2.5mg/kg, i.p., n= 6). The administration of RA was arranged 60 min prior to LPS injections. Subsequently, after 24 hours of supervision, the animals were euthanised via exsanguination with overdosage of sodium pentobarbital and all our efforts aimed to minimise pains of the animals. Meanwhile, all heart samples were collected for histology analyses or immediately frozen in liquid nitrogen, preserved under 80 for future biochemistry examination. Lipopolysaccharide (LPS), Retinoic Acid (RA) and Dexamethasone (Dex) were bought from Sigma (St Louis, MO, USA).

The gene expression dataset GSE44363 related to endotoxemic myocarditis was acquired from the GEO database.26 The GSE44363 data collection, which involved 4 normal and 4 endotoxemic myocardium that were treated for 24 hours with either saline or LPS, was based on wild-type mice. All RNA information of the chosen specimens was acquired for future analysis. The limma package27 was employed to calculate the difference between two groups of patients, and gene screening conditions with p.adj<0.05 and | Log2FC | > 1 were used for filtering DEGs in SIC.

The chemistry structure and SMILES of Retinoic acid was acquired from PubChem web site.28 The target forecast of Retinoic acid was completed via the STITCH data base (http://stitch.embl.de/),29 while the species was limited to Mus musculus.

GO function analysis (CC, BP, and MF) was a potent biological information method to categorize genetic expressing and its performances,30,31 while KEGG pathway analysis was adopted to determine which cellular pathway may participate in the variations of DEGs.32,33 The visualization of GO enriching assay (p.adj<0.05) and KEGG pathway assay (p.adj<0.05) was realized via the R ggplot2 package.

The PPI net of targeted genes was acquired via from STRING 11.0 database,34 with minimal needed interactive score 0.7. The visualization of the PPI net was realized via Cytoscape 3.7.2.35 In the net, nodal points denoted targeted protein, and edges denoted the forecasted or verified mutual effect between protein. Topology analyses of targeted genes were completed via the Cytohubba plug-in of Cytoscape. Targeted protein was subjected to filtration, respectively, as per the BottleNeck, Betweenness, Stress and Radiality subnetworks, which were computed via Cytohubba plug-in. Top 10 genes of every sub-net were searched, and overlapping genes were chosen as critical targets herein.

Another 50 mice were stochastically separated into the following groups: (1) Saline (i.p., n = 10), (2) LPS (10 mg/kg i.p., n = 10), (3) LPS (10 mg/kg) plus RA (1 mg/kg, i.p., n = 10), (4) LPS (10 mg/kg) plus RA (3 mg/kg, i.p., n = 10), (5) LPS (10 mg/kg) plus Dexamethasone (DEX) (2.5 mg/kg, i.p., n = 10). Administration of saline, RA, or DEX was scheduled 60 minutes before LPS injection. The intervention and modeling methods of mice in each group were the same as before. After modeling, the 7-day survival rates of the five groups of mice were observed.

CK-MB (MEIMIAN, China) and LDH (LEAGENE, China) levels in serum were quantified using kits according to the manufacturer's instructions.

Our team prepared samples according to the test kit specifications. The levels of catalase (CAT),36 superoxide dismutase (SOD)37 and GSH/GSSG38 in the heart samples were determined by colorimetry according to the kits in previous studies mentioned above. The results of CAT and SOD were expressed in units of protein per mg (U/mg prot).

Echocardiography was implemented by a Vevo 3100 ultrasonic equipment with a 10-MHz linear array ultrasonic transducer (Fujifilm, VisualSonics, USA) after mice were anesthetized by 1.5% isoflurane. As medial echocardiographic readings were collected from 35 heart cycles, heart function indexes, such as fractional shortening (FS), ejection fraction (EF), etc., were documented.

Overall RNA from all frozen cardiac tissues was abstracted via TRIzol reagent (Invitrogen). Overall RNA (2 g) was converted to cDNA through reverse transcription by cDNA synthesis kit (Takara Clontech, Dalian, Japan). qPCR was completed by toroivd SYBR Green qPCR Master Mix (20 L). The cycling status were stated below: denaturalization under 95 for 60s, 40 cycles under 95 for 10s, 60 for 30s, and 72 for 45s. The RNA quantity was computed via the comparative threshold cycle approach, with every primer customized by Sangon Biotechnology Co., Ltd. (Shanghai, China). Eventually, each primer sequence is presented in Table 1.

Table 1 Primers Used for qPCR of Genes from Mouse

Proteins were abstracted from the entire frozen cardiac tissues via RIPA lysis buffering solution with 1% protease suppressor mixture. The BCA approach was employed to compute the protein level. Equal amounts of proteins were separated by 1015% SDS-PAGE. Samples were moved onto PVDF films, subjected to blockade via 5% dry skimmed milk, and incubated with primary antibodies including anti-ITGAM (Abcam, Ab133357, 1:1000), anti-VCAM1 (Abcam, Ab134047, 1:1000), anti-PPARA (Abcam, Ab61182, 1:1000), anti-IGF1 (Abcam, Ab9572, 1:1000), anti-IL-6 (Abcam, Ab259341, 1:1000), anti-GAPDH (Abcam, Ab181602, 1:2000), anti-phospho-Akt S473 (Cell Signaling Technology, 4060, 1:1000) and anti-Akt (Cell Signaling Technology, 4691, 1:1000) separately, at 4 nightlong. Posterior to the cleaning in TBST, the blots were cultivated with antirabbit or antimouse second antisubstances for 60 min under ambient temperature. Afterwards, the outcomes were identified via the ECL identification reagents. Proteins in Western blot were quantified via Image Lab software. All assays were completed in triplicate.

Cardiac specimens were subjected to 4% neutral PFA fixation, paraffin embedment, and sectioning. For H&E dyeing (Solarbio, Beijing, China), 5 m slices were dyed in hematoxylin for 600 s, and afterwards cleaned and dyed in 0.5% eosin for 300 s. Posterior to the cleaning in water, the samples were subjected to dehydration in 70%, 85%, 95%, and 100% ethyl alcohol and afterwards in xylene. Heart injuries were analyzed by microscopic fields of every tissular specimen, which was stochastically chosen. The morphological status of myofilament and inflammation cell infiltration were evaluated as standard.

For IHC, paraffin slices were subjected to deparaffinization via xylene and subjected to rehydration via the concentration gradient of ethyl alcohol. Subsequently, antigen repair was completed and the specimens were cultivated with anti-CD68 (CST, D4B9C, 1:200) nightlong at 4 . Eventually, the slices were cultivated by an antirabbit EnVisionTM +/HRP reagent for sixty minutes at 37 for the observation via a light microscopy (Nikon, H550L, Tokyo, Japan).

By virtue of the TUNEL approach, heart slices were dyed for identifying DNA fragmentation, which could reflect cell apoptosis. Slices were cultivated in TdT-reaction liquor and the visualization of nuclei was realized via TUNEL reagents (Promega, America) and DAPI nuclear dye. Fluorescent pictures were captured via a Nikon microscopic device. Quantitation of TUNEL-positive cells was completed via identifying the corresponding proportion (%) (green) in several high-power fields (n = 3 slices every mouse strain and treatment group).

The experiment data were studied via GraphPad Prism 8.0. The entire experiment data were described as average SD. Students t-test was used for comparison between the two groups, and the diversities between the groups were compared by two-way ANOVA and corrected by Bonferroni. P < 0.05 had significance on statistics. Survival rate was evaluated by KaplanMeier analysis.

The 2D structure of RA was acquired from PubChem (Figure 1A). An overall 500 genes were identified as targeted genes of RA from the STITCH database. In addition, 1035 DEGs were selected from GSE44363 dataset, and 547 genes were regulated upward, with 488 regulated downward (Figure 1B). By pairing DEGs with RA targets (Figure 1C), 54 genes were chosen as underlying targeted genes in septic cardiac dysfunction. The thermograph of those 54 genes was presented by Figure 1D.

Figure 1 Target genes of RA and DEGs in GSE44363. (A) Chemical structure of Retinoic acid; (B) DEGs in GSE44363 (Upregulated genes were marked in red and downregulated genes were marked in blue). (C) Venn diagram of Retinoic acid target genes and DEGs. (D) Clustered heat map of overlapped genes .

GO analyses of the 54 underlying treatment target genes were completed via the DAVID database. Targeted genes were primarily enriched in the regulation of ossification, myeloid leukocyte differentiation and epithelial cell proliferation in BP enrichment analysis, and they were also enriched in extracellular matrix, collagen-containing extracellular matrix and membrane raft in CC analysis. In MF analysis, they were enriched in glycosaminoglycan binding, heparin binding, and cytokine activity (Figure 2A). The outcome of KEGG pathway enriching analyses revealed that targeted genes were remarkably enriched in the PI3K-Akt signaling pathway, transcriptional misregulation in cancer, and TNF signaling pathway, etc. (Figure 2B and C).

Figure 2 Enrichment Analysis of Overlapped Target. (A) Gene ontology (GO) enrichment analysis for key targets (Top 10 were listed). (B) KEGG pathway enrichment analysis of key targets (Top 10 were listed); the abscissa label represents GeneRatio. (C) KEGG pathway analysis and related genes (Top ten were listed).

The PPI net of aforesaid targeted proteins was established via STRING and the visualization of the network was realized via Cytoscape. The PPI net comprised 54 nodal points and 266 edges (Figure 3A). BottleNeck, Betweenness, Stress and Radiality of targeted protein were computed via topology analyses (Figure 3BE). Top 10 hub nodes of BottleNeck, Betweenness, Stress and Radiality sub-nets were searched, and we discovered 5 overlapping genes: Peroxisome proliferator activated receptor alpha (PPARA), Integrin Subunit Alpha M (ITGAM), Vascular cell adhesion molecule-1 (VCAM1), Insulin-like growth factor 1 (IGF-1), and Interleukin-6 (IL-6) (Figure 3F).

Figure 3 PPI network construction. (A) PPI network construction of overlapped genes (Retinoic acid target genes and DEGs). (BE) Top 10 genes with the highest BottleNeck, Betweenness, Stress and Radiality. (F) Venn diagram summarizing overlapped genes in four sections.

Our team studied the role of RA in survival condition by intraperitoneally injecting male C57BL/6 mice with LPS (10mg/kg), LPS (10 mg/kg) plus RA (1 mg/kg), LPS (10 mg/kg) plus RA (3 mg/kg), LPS (10 mg/kg) plus DEX (2.5 mg/kg) or an equal volume of saline for 7 days. As presented in Figure 4A, our team computed the 7-day survival rate in the five groups below: the Sham group, LPS group, LPS + RA (1 mg/kg) group, LPS + RA (3 mg/kg) group, LPS + DEX (2.5 mg/kg) group. The 7-day survival rate in the Sham group was nearly a hundred percent, whereas 7 days posterior to LPS treatment, the survival rate of LPS group dropped notably to 40%. RA (1 mg/kg) pretreatment enhanced the survival rate to 50% in LPS-exposed animals. RA (3 mg/kg) pretreatment elevated the survival rate to 70% in LPS-exposed animals. DEX (2.5 mg/kg) pretreatment elevated the survival rate to 60% in LPS-exposed animals.

Figure 4 Determine the effective concentration of RA. (A) Survival curve of mice treated with saline, LPS (10 mg/kg), LPS (10 mg/kg) plus RA (1 mg/kg), LPS (10 mg/kg) plus RA (3 mg/kg), LPS (10 mg/kg) plus DEX (2.5 mg/kg). Observe and record the mortality of mice within 1 week (n = 10) .(B and C) The serum levels of CK-MB and LDH in mice treated with saline, LPS (10 mg/kg), LPS (10 mg/kg) plus RA (1 mg/kg), LPS (10 mg/kg) plus RA (3 mg/kg), LPS (10 mg/kg) plus DEX (2.5 mg/kg) for 24h were determined. *P<0.05, **P < 0.01, ***P < 0.001 vs LPS. ns: no significant difference.

Since LDH and CK-MB are sensitive biomarkers of cardiac injury, our team measured LDH and CK-MB levels in serum. We found that using RA (1 mg/kg), RA (3 mg/kg) and DEX (2.5 mg/kg) significantly reduced the level of CK-MB in LPS-injected mice, and the administration of RA (3 mg/kg) displayed the most significant effect (Figure 4B). Meanwhile, the administration of RA (3 mg/kg) and DEX (2.5 mg/kg) significantly reduced LDH levels in LPS-injected mice. However, the administration of RA (1 mg/kg) also reduced LDH in LPS-injected mice, whereas it was not statistically significant (Figure 4C). As high-dose group showed a more significant efficacy when it came to the improvement of mortality and myocardial injury in mice, the dose of RA was 3 mg/kg in the subsequent experiments.

As shown in Figure 5AC, after LPS treatment, SOD, CAT, GSH/GSSG in the heart tissue of mice in the model group were significantly lower than those in the Sham group (P < 0.05), while SOD, CAT, GSH/GSSG in the heart tissue of the RA group were significantly higher than those in the model group (P < 0.05). The activation of inflammatory response marks one of the most essential pathology variations in sepsis-caused cardiac muscle injury. Hence, our team studied the inflammatory cell infiltration and the mRNA expression of proinflammatory cytokines in all groups. As shown in Figure 5DF, qRT-PCR results displayed the favorable effect of RA on heart inflammatory events induced by LPS, as proven by the reduced mRNA contents of TNF-, IL-1 and IL-6 in myocardium tissues. By virtue of the echocardiographic method, our team explored heart functions in 3 groups. RA pretreatment reinforced ejection fraction and fraction shortening in LPS-exposed animals (Figure 5G and 5H).

Figure 5 RA suppressed oxidative stress, cardiac inflammation, and cardiac injury in LPS-treated mice. (AC) SOD, CAT, GSH/GSSG levels in myocardial tissue of each group. (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001 vs LPS. (DF) The mRNA levels of IL-1, IL-6 and TNF- in myocardial tissues of each group (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001 vs LPS. (G and H) Effects of saline, LPS and LPS+RA on left ventricle fractional shortening and left ventricle ejection fraction (n = 6). (IJ) Representative images of the morphological analysis and inflammatory cells infiltration as reflected by the H&E staining, and immunohistochemistry staining for CD68 protein.(K) TUNEL assay was used to detect apoptosis of cardiac tissue in each group.

The myocardium slices were dyed by H&E to evaluate the heart muscle injury and inflammatory cell infiltration. Histological features of heart damage, such as evident capillary congestion, interstitial tissue oedema, and infiltration of massive inflammation cells, were identified in the LPS group. However, in the LPS + RA group, the myocardium fibers registered obvious striation and little inflammatory infiltration was detected in heart muscle tissues (Figure 5I). IHC dyeing revealed that the infiltrative activities of CD68-labeled macrophages, which were caused by LPS, were inhibited by Retinoic acid (Figure 5J). Furthermore, the cardiac injury was evaluated in 3 groups. Tunel dyeing outcomes revealed that the LPS + RA group exhibited less programmed cell death in contrast to the LPS group (Figure 5K). Taken together, those data revealed that Retinoic acid could ameliorate oxidative stress, cardiac inflammation, cardiac injury and heart functions in septic mice.

To verify the network pharmacological forecast of Retinoic acid in LPS-treated mice, we performed qRT-PCR and WB observation to calculate the PPARA, ITGAM, VCAM-1, IGF-1, and IL-6 levels in healthy cardiac samples and cardiac samples from the LPS group and LPS + RA group. Posterior to the normalization with GAPDH, the expressing levels of ITGAM, VCAM-1, and IL-6 were remarkably elevated in the LPS group in contrast to the Sham group, whereas the expressing levels of those biomarkers were remarkably decreased (P< 0.05) in the RA-exposed group in contrast to the LPS group. Meanwhile, the expression levels of PPARA and IGF-1 were remarkably reduced in the LPS group in contrast to the Sham group, whereas the expressing levels of them were remarkably increased (P < 0.05) in the RA-exposed group (Figures 5E and 6A). Then, Western blot analyses verified that Retinoic acid markedly diminished the ITGAM, VCAM-1, and IL-6 protein levels compared with the LPS group, and LPS markedly decreased the protein levels of PPARA and IGF-1. In addition, RA reversed LPS-induced PPARA and IGF-1 inhibition as expected (Figure 6BF). Therefore, those outcomes revealed that RA might suppress the stimulation of inflammation reactions and engage in the progress of SIC by means of the aforementioned molecules.

Figure 6 RA modulated PPARA, ITGAM, VCAM-1, and IGF-1 in hearts of LPS-treated mice. (A) The mRNA levels of ITGAM, VCAM-1, PPARA and IGF-1 in myocardial tissues of each group (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001 vs LPS. (BF) The protein levels of ITGAM, VCAM-1, PPARA, IGF-1, and IL-6 in myocardial tissues of each group (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001 vs LPS.

In order to further verify the network pharmacological prediction of RA in lipopolysaccharide-induced cardiac dysfunction, Western blot analysis was performed to detect the phosphorylation level of Akt. The results showed that RA could restore the expression of P-Akt in LPS-treated mouse heart tissues (P < 0.05) (Figure 7).

Figure 7 RA affects the expression of p-Akt in hearts of LPS treated mice. (A) Representative Western blot images of p-Akt, Akt. (B) Densitometric quantification analysis of the protein expression levels of p-Akt and Akt in mice. **P < 0.01, ***P < 0.001 vs LPS.

The definition for sepsis from the third international consensus states that sepsis is a lethal organ function disorder induced by an aberrant reaction to infections.39 Cytokines, especially IL1, IL6, and TNF, are major contributors in the initiation of SIC.40 It has been demonstrated that when endotoxin like LPS binds to the receptor TLR4 expressed on cardiomyocytes and macrophages, these cells can release massive inflammatory mediators, such as MCP-1, GM-CSF, IL-1, IL-1, IL-6, IL-7, IL-8, and IL-12.41

Retinoic acid (RA) has been reported to reduce the levels of circulation endotoxin and ameliorate survival in endotoxaemic rats.42 Furthermore, RA was a powerful derivant of vitamin A. In previous studies, administrating vitamin A to infants and minors could decrease the risks of septic diseases, immune deficiencies and inflammatory events in endemic regions with deficient vitamin.17,43,44 Eriksson et al also demonstrated that vitamin A administered before a E. coli endotoxin infusion modified the harmful events on heart-lung systems which was caused by such LPS.45 Pretreating with vitamin A counteracted the role of endotoxin in mean arterial pressure (MAP) and cardiac index (CI). Therefore, exploring the mechanism by which RA fights against SIC is quite pregnant.

Network pharmacology method is a comparatively new way to investigate the treatment potency and potential causal links of medicines on the foundation of the net of medicines and targets.46 Herein, our team utilized network pharmacology method to explore the treatment targets participating in the RA healing of SIC. We revealed that RA exposure could elevate survival rate and heart functions of LPS-induced mice while inhibiting inflammatory cytokines and oxidative stress of cardiac muscles. Furthermore, the treatment of RA reversed the production of PPARA, ITGAM, VCAM-1, IGF-1, and IL-6 in LPS-induced SIC.

PPARs are vital targets for approved and experiment medicines in substantial clinical indications, such as metabolism and inflammation illnesses.4749 The expression of PPARA is extensive in our bodies (particularly in hearts, kidneys, and livers), as an important regulator in the heart after LPS administration.50 Described that heart PPARA expression was imperative for protecting against sepsis-triggered heart damage.51 The 3 PPAR sub-groups, PPAR, PPAR, and PPAR/ generate heterodimers with their obligatory dimer partner RXR,52 and RA modulates genetic expression straightly via binding to a heterodimer of the RARs and RXRs, which are capable of binding to RAREs in the modulatory area of the targeted genes.17 These results suggest that altered RA-mediated PPARA expression might be vital for sepsis-associated end-organ damage and function disorder, particularly in cardiac tissues.

Integrin subunit alpha M (ITGAM) was discovered to be highly expressed in adults and sepsis of the newborn.53,54 A past research unraveled that ITGAM primarily facilitated septic development via fostering the nucleus, cytoplasm movement and stimulating the releasing of HMGB1.55 ITGAM block antibodies or suppressors could defend mice against the fatalness related to LPS and microbe sepsis.56 ITGAM, namely CD11b, regulates the activation, adhesion, and migration of leucocytes from blood to injury sites.57 Vitamin A and its active metabolite retinoic acid (RA) are essential for the development and function of the immune system. Recent studies have also indicated that vitamin A stimulates the development of CD11b+ dendritic cells, and affects the generation of a specific niche that drives CD11b+ dendritic cells (CD11b+ DC) differentiation.5861 Thus, we speculated that RA might exert an anti-SIC effect via preventing the ITGAM-associated leukocyte recruitment to inflamed tissues.

The vascular cell adhesion molecule (VCAM-1), a heterodimeric molecule expressed on the surface of leukocytes, is induced in inflammatory stimulation.46,6264 Mice deficient of endothelial selectins exhibited increased survival in an animal model of sepsis.65,66 Furthermore, increased serum content of VCAM1 was a superior predicting factor for sepsis-induced brain diseases in adult community-onset sepsis on admission.67 Recently, Moser J et al have identified that RIG-I, a new modulator of endothelium pro-inflammation stimulation functioning in parallel with TLR4, can regulate the expressing of VCAM-1 in reaction to LPS exposure.68 Gille et al reported that pretreatment with all-trans-retinoic acid prevented the TNFa-mediated VCAM-1 induction.69 In the present study, the experiment outcomes revealed the suppressive role of RA in VCAM-1 generation.

The insulin-like growth factor-1 (IGF-1), a hormone with an insulin-alike architecture, is the main mediating factor of growing hormone.70 Additionally, studies have demonstrated that oxidative stress regulates the level of IGF-1, which is reduced in the acute phase of critical patients blood samples,71,72 and that IGF-1 can facilitate the growth and repairment of hearts.73 Furthermore, IGF-1 may defend our hearts against sepsis-triggered myocarditis.7476 In another study, the researchers found that RA increased the production of THREE-DIMENSIONAL human dermal equivalents (HDEs) IGF1 and IGF2.77 The WB outcome revealed that RA restored the expressing of IGF1 in LPS-induced cardiac dysfunction.

The Phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway is a classic signaling pathway. It plays an important role in regulating cell growth, proliferation, differentiation, metabolism, cytoskeletal reorganization, autophagy and apoptosis.7880 Various growth factors and cytokines activate the PI3K/Akt signaling pathway, which ultimately phosphorylates Akt. Substantial studies have revealed that activated Akt1 has cardioprotective effects.8183 A number of studies have found that the apoptosis of myocardial cells in Akt2 knockout mice is more serious than that in normal mice during myocardial ischemia, which reveals that Akt2 can also reduce the apoptosis of myocardial cells and protect the heart.84 Therefore, the activation of the PI3K/Akt signaling pathway can reduce cardiac injury and protect cardiac function through various ways. By analyzing the potential target of RA-treated lipopolysaccharide-induced cardiac dysfunction, we discovered that the PI3K/Akt signaling pathway was the key pathway in the RA treatment of lipopolysaccharide-induced cardiac dysfunction. After experimental verification, we found that RA could restore the activation of the PI3K/Akt signaling pathway in the heart tissues of LPS-treated mice.

Hence, our team estimate that RA might be an anti-inflammation agent in SIC. Nevertheless, there were certain deficiencies in our research. First, as the experimental subjects were merely mice, future researches have to investigate the roles of lipopolysaccharide-induced cardiac dysfunction in human. Furthermore, the effects of the 5 key targets of RA on LPS-induced cardiac dysfunction in mice require further exploration so as to unravel the corresponding mechanisms underneath.

In the present research, we analyzed the cellular components, biological processes, functions, and relevant pathways of retinoic acid, as well as its molecular effects on lipopolysaccharide-induced cardiac dysfunction in mice through extensive bioinformatics analysis. Using the Cytohubba software, we successfully identified five potential key therapeutic targets. In addition, by regulating 5 survival-related key therapeutic targets and a key pathway, PI3K-Akt signaling pathway, our team confirmed that Retinoic acid could be a potential therapeutic drug for lipopolysaccharide-induced cardiac dysfunction.

SIC, sepsis-induced cardiomyopathy; CC, cellular component; DEGs, differentially expressed genes; LPS, lipopolysaccharide; RA, retinoic acid; MF, molecular function; FS, fractional shortening; IHC, immunohistochemistry; TdT, terminal deoxynucleotidyl transferase; RAREs, RA response elements; MAP, Mean Arterial Pressure; PPI, proteinprotein interaction; BP, biological process; CI, cardiac index; PPARs, peroxisome proliferator-activated receptors; RXR, retinoid X receptor; ATRA, all-trans retinoic acid; EF, ejection fraction; RXRs, retinoid X receptors; ITGAM, integrin subunit alpha M; HDEs, human Dermal equivalents; LN, liquid nitrogen; SMILES, simplified molecular input line entry specification; H&E, hematoxylin and eosin; WB, Western blot; RIG-I, retinoic acid inducible gene-I; RARs, RA nuclear receptors.

The datasets used and/or analyzed during the current study are available from the corresponding author and GEO database (https://www.ncbi.nlm.nih.gov/geo).

According to the Regulations and Rules of Guidelines for Ethical Review of the Welfare in Laboratory Animals (2018), each animal assay was accepted by the Animal Experimentation Ethics Committee of Wenzhou Medical University (Approval Ethical Inspection ID: WYYY-AEC-2021-301).

We are grateful for the experimental instruments provided by the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.

Xi Wang and Chang Kong share first authorship. X. Wang and H. Tang conceived and designed the experiments. Ch. Kong, X. Wang and P. Liu executed the experiments and analyzed the samples. X. Wang, B. Zhou and W. Geng analyzed the data. X. Wang and Ch. Kong wrote the first version of the manuscript. All authors interpreted the data, critically revised the manuscript, and approved the final version of the manuscript. All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; agreed to submit to the current journal; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.

We acknowledge the funding received from the Natural Science Foundation of China (Grant No. 81774109 and 81973620) and Wenzhou Municipal Science and Technology Bureau (ZY2019015).

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

1. Qian M, Lou Y, Wang Y, et al. PICK1 deficiency exacerbates sepsis-associated acute lung injury and impairs glutathione synthesis via reduction of xCT. Free Radic Biol Med. 2018;1(18):2334. doi:10.1016/j.freeradbiomed.2018.02.028

2. Fernando SM, Rochwerg B, Seely A. Clinical implications of the third international consensus definitions for sepsis and septic shock (Sepsis-3). CMAJ. 2018;36(190):E10589. doi:10.1503/cmaj.170149

3. Hollenberg SM, Singer M. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol. 2021;6(18):424434. doi:10.1038/s41569-020-00492-2

4. Weiss SL, Cvijanovich NZ, Allen GL, et al. Differential expression of the nuclear-encoded mitochondrial transcriptome in pediatric septic shock. Crit Care. 2014;6(18):623. doi:10.1186/s13054-014-0623-9

5. Meyer NJ. Finding a needle in the haystack: leveraging bioinformatics to identify a functional genetic risk factor for sepsis death. Crit Care Med. 2015;1(43):242243. doi:10.1097/CCM.0000000000000664

6. Burnham KL, Davenport EE, Radhakrishnan J, et al. Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia. Am J Respir Crit Care Med. 2017;3(196):328339. doi:10.1164/rccm.201608-1685OC

7. Scicluna BP, van Vught LA, Zwinderman AH, et al. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir Med. 2017;10(5):816826.

8. Jia L, Wang Y, Wang Y, et al. Heme oxygenase-1 in macrophages drives septic cardiac dysfunction via suppressing lysosomal degradation of inducible nitric oxide synthase. Circ Res. 2018;11(122):15321544. doi:10.1161/CIRCRESAHA.118.312910

9. Smeding L, Plotz FB, Groeneveld AB, et al. Structural changes of the heart during severe sepsis or septic shock. Shock. 2012;5(37):449456. doi:10.1097/SHK.0b013e31824c3238

10. Vanasco V, Saez T, Magnani ND, et al. Cardiac mitochondrial biogenesis in endotoxemia is not accompanied by mitochondrial function recovery. Free Radic Biol Med. 2014;77:19. doi:10.1016/j.freeradbiomed.2014.08.009

11. Wagner S, Schurmann S, Hein S, et al. Septic cardiomyopathy in rat LPS-induced endotoxemia: relative contribution of cellular diastolic Ca(2+) removal pathways, myofibrillar biomechanics properties and action of the cardiotonic drug levosimendan. Basic Res Cardiol. 2015;5(110):507. doi:10.1007/s00395-015-0507-4

12. Schilling J, Lai L, Sambandam N, et al. Toll-like receptor-mediated inflammatory signaling reprograms cardiac energy metabolism by repressing peroxisome proliferator-activated receptor gamma coactivator-1 signaling. Circ Heart Fail. 2011;4(4):474482. doi:10.1161/CIRCHEARTFAILURE.110.959833

13. Ziolo MT, Katoh H, Bers DM. Expression of inducible nitric oxide synthase depresses beta-adrenergic-stimulated calcium release from the sarcoplasmic reticulum in intact ventricular myocytes. Circulation. 2001;24(104):29612966. doi:10.1161/hc4901.100379

14. Alvarez S, Vico T, Vanasco V. Cardiac dysfunction, mitochondrial architecture, energy production, and inflammatory pathways: interrelated aspects in endotoxemia and sepsis. Int J Biochem Cell Biol. 2016;81(Pt B(81)):307314. doi:10.1016/j.biocel.2016.07.032

15. Nurrahmah QI, Madhyastha R, Madhyastha H, et al. Retinoic acid abrogates LPS-induced inflammatory response via negative regulation of NF-kappa B/miR-21 signaling. Immunopharmacol Immunotoxicol. 2021;3(43):299308. doi:10.1080/08923973.2021.1902348

16. Li S, Lei Y, Lei J, et al. All-trans retinoic acid promotes macrophage phagocytosis and decreases inflammation via inhibiting CD14/TLR4 in acute lung injury. Mol Med Rep. 2021;24(6). doi:10.3892/mmr.2021.12508

17. Austenaa LM, Carlsen H, Hollung K, et al. Retinoic acid dampens LPS-induced NF-kappaB activity: results from human monoblasts and in vivo imaging of NF-kappaB reporter mice. J Nutr Biochem. 2009;9(20):726734. doi:10.1016/j.jnutbio.2008.07.002

18. Martire-Greco D, Landoni VI, Chiarella P, et al. all-trans-retinoic acid improves immunocompetence in a murine model of lipopolysaccharide-induced immunosuppression. Clin Sci. 2014;5(126):355365. doi:10.1042/CS20130236

19. Tang M, Xie X, Yi P, et al. Integrating network pharmacology with molecular docking to unravel the active compounds and potential mechanism of simiao pill treating rheumatoid arthritis. Evid Based Complement Alternat Med. 2020;2020:5786053. doi:10.1155/2020/5786053

20. Liu Z, Huo JH, Dong WT, et al. A study based on metabolomics, network pharmacology, and experimental verification to explore the mechanism of qinbaiqingfei concentrated pills in the treatment of mycoplasma pneumonia. Front Pharmacol. 2021;12:761883. doi:10.3389/fphar.2021.761883

21. Hopkins AL. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 2008;11(4):682690. doi:10.1038/nchembio.118

22. Recanatini M, Cabrelle C. Drug research meets network science: where are we? J Med Chem. 2020;16(63):86538666. doi:10.1021/acs.jmedchem.9b01989

23. Hopkins AL. Network pharmacology. Nat Biotechnol. 2007;10(25):11101111. doi:10.1038/nbt1007-1110

24. Tao W, Xu X, Wang X, et al. Network pharmacology-based prediction of the active ingredients and potential targets of Chinese herbal Radix Curcumae formula for application to cardiovascular disease. J Ethnopharmacol. 2013;1(145):110. doi:10.1016/j.jep.2012.09.051

25. Fang J, Liu C, Wang Q, et al. In silico polypharmacology of natural products. Brief Bioinform. 2018;6(19):11531171. doi:10.1093/bib/bbx045

26. Barrett T, Wilhite SE, Ledoux P, et al. NCBI GEO: archive for functional genomics data setsupdate. Nucleic Acids Res. 2013;41(Database issue(41)):D9915. doi:10.1093/nar/gks1193

27. Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;7(43):e47. doi:10.1093/nar/gkv007

28. Kim S, Chen J, Cheng T, et al. PubChem in 2021: new data content and improved web interfaces. Nucleic Acids Res. 2021;49(D1(49)):D138895. doi:10.1093/nar/gkaa971

29. Szklarczyk D, Santos A, von Mering C, et al. STITCH 5: augmenting protein-chemical interaction networks with tissue and affinity data. Nucleic Acids Res. 2016;44(D1(44)):D3804. doi:10.1093/nar/gkv1277

30. Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000;1(25):2529. doi:10.1038/75556

31. Li S, Li L, Meng X, et al. DREAM: a database of experimentally supported protein-coding RNAs and drug associations in human cancer. Mol Cancer. 2021;1(20):148. doi:10.1186/s12943-021-01436-1

32. Altermann E, Klaenhammer TR. PathwayVoyager: pathway mapping using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. BMC Genom. 2005;6:60. doi:10.1186/1471-2164-6-60

33. Backes C, Keller A, Kuentzer J, et al. GeneTrailadvanced gene set enrichment analysis. Nucleic Acids Res. 2007;35(Web Server issue(35)):W18692. doi:10.1093/nar/gkm323

34. Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1(47)):D60713. doi:10.1093/nar/gky1131

35. Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;11(13):24982504. doi:10.1101/gr.1239303

36. Liu C, He D, Zhao Q. Licoricidin improves neurological dysfunction after traumatic brain injury in mice via regulating FoxO3/Wnt/beta-catenin pathway. J Nat Med. 2020;4(74):767776. doi:10.1007/s11418-020-01434-5

37. Zhou Q, Wang X, Shao X, et al. tert-butylhydroquinone treatment alleviates contrast-induced nephropathy in rats by activating the Nrf2/Sirt3/SOD2 signaling pathway. Oxid Med Cell Longev. 2019;2019:4657651. doi:10.1155/2019/4657651

38. Deng C, Zhang B, Zhang S, et al. Low nanomolar concentrations of Cucurbitacin-I induces G2/M phase arrest and apoptosis by perturbing redox homeostasis in gastric cancer cells in vitro and in vivo. Cell Death Dis. 2016;7:e2106. doi:10.1038/cddis.2016.13

39. Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;8(315):801810. doi:10.1001/jama.2016.0287

40. Drosatos K, Lymperopoulos A, Kennel PJ, et al. Pathophysiology of sepsis-related cardiac dysfunction: driven by inflammation, energy mismanagement, or both? Curr Heart Fail Rep. 2015;2(12):130140. doi:10.1007/s11897-014-0247-z

41. Rossol M, Heine H, Meusch U, et al. LPS-induced cytokine production in human monocytes and macrophages. Crit Rev Immunol. 2011;5(31):379446. doi:10.1615/critrevimmunol.v31.i5.20

42. Drott PW, Meurling S, Kulander L, et al. Effects of vitamin A on endotoxaemia in rats. Eur J Surg. 1991;10(157):565569.

43. Slade E, Tamber PS, Vincent JL. The surviving sepsis campaign: raising awareness to reduce mortality. Crit Care. 2003;1(7):12. doi:10.1186/cc1876

44. Carcillo JA. Reducing the global burden of sepsis in infants and children: a clinical practice research agenda. Pediatr Crit Care Med. 2005;3(Suppl(6)):S15764. doi:10.1097/01.PCC.0000161574.36857.CA

45. Eriksson M, Lundkvist K, Drott P, et al. Beneficial effects of pre-treatment with vitamin A on cardiac and pulmonary functions in endotoxaemic pigs. Acta Anaesthesiol Scand. 1996;5(40):538548. doi:10.1111/j.1399-6576.1996.tb04485.x

Here is the original post:
Therapeutic Effects of Tretinoin | JIR - Dove Medical Press

Posted in Genetic Medicine | Comments Off on Therapeutic Effects of Tretinoin | JIR – Dove Medical Press

Manor College 75 | Marianne Tracey ’61, Prominent in the Field of Psychiatry – Manor College

Dr. Marianne Tracey came to Manor College from Cathedral High School.

Dr. Marianne Tracey was born in Trenton, New Jersey, and upon graduating from Cathedral High School enrolled at Manor College. She earned her associates degree at Manor College in 1961. From there, she had a distinguished journey as a lifelong learner, professor, and researcher. She went on to graduate from several prominent colleges and universities, and her work in the field of intellectual disabilities was especially noteworthy.

She was named the Superintendent of the Edison Development Center for the Developmental Disabled/Behavioral Males, according to her memorial biography.

In this capacity, the memorial states, she determined that what appeared to be behavioral problems in this population, were in fact often symptoms of genetic, neurological, endocrinologic and psychiatric disorders, a radical idea for that time (this is now called Dual Diagnosis). She began working on creating a medical and psychiatric liaison service with what was then the University of Medicine and Dentistry of New Jersey (UMDNJ). In 1988, she created what has become the Center of Excellence for the Mental Health Treatment of Persons with Intellectual Disabilities and Autism Spectrum Disorders of UMDNJ-SOM (later Rowan University-SOM).

For 30 years, the Center provided psychiatric consultation to New Jerseys Development Centers and outpatient services to persons with Intellectual Disabilities and Autism Spectrum Disorders, based not on behavioral problems but upon psychiatric diagnosis, a pioneering approach at that time. The clinic served patients of all ages and in all living situations in the seven southern counties of New Jersey; its successor organization continues to serve outpatients.

In addition to her professional education, the memorial also notes that Dr. Tracey pursued many other interests. She attended Simca Cuisine School in France in 1982 and New York Cooking School in 1983 and for a number of years was the owner and operator of the Newtown Cheese Shop in Newtown, PA. This is where she lived for more than 40 years until she passed in January 2022. In her spare time, she enjoyed travel and visited many interesting places throughout the world.

Manor College is honored to feature Dr. Tracey as an alumni of the college. Her dedication to the field of psychiatry and her love of lifelong learning is a testimony to the values we have sought to instill in our students over the past 75 years.

Basileiad Library on Display

The Manor College Foxtrotters?

The Blue Jays Best Shooter

Follow this link:
Manor College 75 | Marianne Tracey '61, Prominent in the Field of Psychiatry - Manor College

Posted in Genetic Medicine | Comments Off on Manor College 75 | Marianne Tracey ’61, Prominent in the Field of Psychiatry – Manor College

Ethical Considerations in Precision Medicine and Genetic Testing in Internal Medicine Practice: A Position Paper From the American College of…

  1. Ethical Considerations in Precision Medicine and Genetic Testing in Internal Medicine Practice: A Position Paper From the American College of Physicians | Annals of Internal Medicine  Annals of Internal Medicine
  2. Genetic Testing Should Be Guided by Patients' Best Interests, ACP Position Paper Says  GenomeWeb
  3. ACP offers guidance on the ethical use of genetic testing and precision medicine  EurekAlert
  4. The Ethical Responsibilities of Precision Medicine According to ACP  MD Magazine
  5. ACP offers guidance on the ethical use of genetic testing and precision medicine | ACP Newsroom | ACP  American College of Physicians
  6. View Full Coverage on Google News

Original post:
Ethical Considerations in Precision Medicine and Genetic Testing in Internal Medicine Practice: A Position Paper From the American College of...

Posted in Genetic Medicine | Comments Off on Ethical Considerations in Precision Medicine and Genetic Testing in Internal Medicine Practice: A Position Paper From the American College of…

Startup aiming to push boundaries of gene therapy nets $55M in seed cash – MedCity News

Genetic medicines have a capacity problem. The engineered viruses used to deliver them have limited room for their genetic cargo, which in turn limits the way diseases can be treatedif they can be treated at all. A new biotech company named Replay has assembled a suite of technologies that could enable it to deliver big genes or even multiple genes, and it has emerged with $55 million to advance its research.

The seed round announced Monday was led by KKR & Co. and OMX Ventures.

The delivery vehicle of choice for many experimental genetic medicines is the adeno-associated virus (AAV), which can be engineered to ferry DNA to target cells. The capacity of AAV is just under 5 kilobases (kb). San Diego-based Replay claims it can achieve a payload capacity up to 30 times greater. It aims to do so with its synHSV technology, which employs an engineered herpes simplex virus (HSV). In addition, Replays toolkit includes technologies that enable it to efficiently write its big genes and big DNA, and a technology that can produce off-the-shelf therapies.

The capacity limitation of AAV is apparent in the research of genetic medicines for Duchenne muscular dystrophy. Sarepta Therapeutics, Solid Biosciences and Pfizer have reached clinical testing with their respective gene therapies, each one engineered to deliver a functioning version of the gene needed to treat the inherited muscle-wasting disorder. But the genes that produce the key protein at the root of Duchenne are big, so the therapies are comprised of micro versions of the gene small enough to fit on an AAV vector.

Duchenne is one of the disease targets of Replay. The therapy in development for that muscle-weakening disorder is 14 kb, according to the companys website. But Replay wont be going directly head to head against the field of potential gene therapies for Duchenne. Under Replays business model, the various technologies it owns are developed in a disease-agnostic way. When Replay identifies an area that can specifically be addressed by one or more of its technologies, it forms a product company to pursue that area. The Duchenne research is housed in one such product company.

Technology and product development have different talent requirements, timelines, costs and cultures, Replay CEO and co-founder Lachlan MacKinnon said in a prepared statement. By separating technology development from product development, we have generated a model to accommodate these differences. Our ability to write and deliver big DNA has the potential to disrupt many areas of genomic medicine.

Replay says it has formed five product companies to date. In the eye, one company is focused on retinitis pigmentosa, a group of rare retinal disorders that leads to degeneration of photoreceptors. The Replay website lists two gene therapy constructs for retinitis pigmentosa: one is 7 kb and the other is 9 kb. A Replay skin product company is developing a treatment for dystrophic epidermolysis bullosa, an inherited disorder that leads to extremely fragile skin that is prone to widespread blistering. The experimental therapy of that company is 19.2 kb. Replays brain product company has the biggest of its genetic medicines in development, a 40 kb therapy for Parkinsons disease. A fifth product company is focused on enzyme writing.

There are other startups that, like Replay, are turning to AAV alternatives in the quest for better genetic medicines. Last month, Philadelphia-area startup Code Bio closed a $75 million Series A round of funding to support the development of synthetic DNA-based therapies for two lead indications, Duchenne and type 1 diabetes.

The new round of financing for Replay included participation from Artis Ventures, Lansdowne Partners, SALT, DeciBio Ventures and Axial.

Photo: iLexx, Getty Images

See the rest here:
Startup aiming to push boundaries of gene therapy nets $55M in seed cash - MedCity News

Posted in Genetic Medicine | Comments Off on Startup aiming to push boundaries of gene therapy nets $55M in seed cash – MedCity News

SalioGen Therapeutics Strengthens its Leadership Team to Advance its Gene Coding Platform – BioSpace

CAMBRIDGE, Mass., July 20, 2022 (GLOBE NEWSWIRE) -- SalioGen Therapeutics, a privately held biotechnology company developing Gene CodingTM, a new category of genetic medicine, today announced the expansion of its leadership team with the addition of five highly accomplished scientists, physicians and biotech industry leaders. The companys new appointments include:

As we continue to build on our foundational scientific discoveries and propel our research and development activities toward the clinic, we welcome Cathryn, Pat, Joe, Feng and Oleg to help drive our progress. The additions of these esteemed experts, each distinguished in their respective specialties, will serve to accelerate our growth by supporting core R&D activities, clinical preparations, and quality and operational needs, said Ray Tabibiazar, M.D., chief executive officer and chairman of SalioGen. We look forward to benefitting from their leadership as they help to maximize the potential impact of Gene Coding not only on the genetic medicines industry, but on patients around the world.

Cathryn Clary, M.D., MBA previously served as Senior Vice President of Medical at SSI Strategy, a medical consulting firm. She also served as the acting Chief Medical Officer at clinical-stage gene therapy company Solid Biosciences. Dr. Clary served as both Global Head of Policy and Patient Affairs in the Chief Medical & Patient Safety Office, as well as Chief Scientific Officer and Head of U.S. Medical Affairs and Clinical Development in the General Medicines Division at Novartis. Prior to her experience at Novartis, she served in several executive roles at Pfizer, whereshe was responsible for the medical aspects of Zoloft worldwide, and ultimately became the SVP of US Medical Affairs for the entire Pfizer portfolio across multiple therapeutic areas.

Pat Sacco is highly experienced as a biotechnology and life sciences executive in technical and general operations. Most recently, as an independent consultant, he has worked with a number of advanced therapy medicinal product (ATMP) biotech companies, CDMOs, and specialized program management clients. Previously, he was the Senior Vice President of Technical Operations at both Translate Bio and Shire (now Takeda), and prior to that held roles of increasing responsibility at Wyeth Biopharma (formerly Genetics Institute) and Genzyme. He has contributed to the development, manufacturing, and commercialization of the enzyme replacement therapies REPLAGAL, ELAPRASE, and VPRIV.

Joe Senn, Ph.D. has experience with nearly all therapeutic modalities, including small molecules, biologics, antisense, gene editing and mRNA therapeutics. He most recently served as the Vice President of Nonclinical Development at Moderna Therapeutics for eight years, where he and his team were responsible for progressing over 40 candidates into the clinic. Prior to Moderna, Dr. Senn served as Site Head for Drug Safety at Takeda Pharmaceuticals, where he oversaw development of all immunology products across the portfolio.

Feng Yao, Ph.D. is the inventor of Invitrogen/Thermo Fisher Scientifics T-Rex tetracycline gene switch, a powerful and specific mammalian transcription gene switch. Using this technology, Dr. Yao has established several unique approaches for the genetic engineering of novel recombinant viruses for use in clinical applications across infectious diseases, cancers and neural regeneration. His T-REx technology is widely used and referenced in publications and patent applications, including productions of several FDA approved antibody therapeutics and novel COVID-19 viral vector-based vaccine candidates developed by AstraZeneca and Johnson and Johnson. Before joining SalioGen Therapeutics, Dr. Yao was an Associate Professor of Surgery at the Brigham and Womens Hospital and Harvard Medical School.

Oleg Iartchouk, Ph.D. has built and led multiple genomics technologyteams that werepart ofstartup and large global pharmaceutical and biotechnologycompanies.Prior to joining SalioGen, Dr. Iartchouk served as the Global Head of the global Genomics Platform Group at the Novartis Institute for Biomedical Research, which established genomics applications several fields to advance Novartiss cell and gene therapy portfolioincluding CAR-T cell(Kymriah) and gene (Zolgensma) therapies.Dr. Iartchouk also built the Biomarkers Discovery group at the clinical diagnostics company Natera and the Applied Genomics team at Sanofi Oncology.

About SalioGen TherapeuticsSalioGen Therapeutics has launched Gene CodingTM, a genetic medicine platform, to develop durable, broadly applicable genetic medicines, using its Exact DNA Integration TechnologyTM (EDITTM) platform. EDIT is based on the novel mammal-derived genomic engineering tool, for use in potentially curative genetic medicines. SalioGen is focused on developing therapies for more patients with inherited diseases that are beyond what is addressable with current technologies, initially focusing on inherited macular disorders and inherited lipid disorders.

For more information, please visit http://www.saliogen.com, or follow us on Twitter and LinkedIn.

Forward-Looking StatementsThis press release contains forward-looking statements. Words such as may, believe, will, expect, plan, anticipate, estimate, intend and similar expressions (as well as other words or expressions referencing future events, conditions or circumstances) are intended to identify forward-looking statements. Forward-looking statements are based upon current estimates and assumptions and include statements regarding the additions of the esteemed experts serving to accelerate SalioGens growth by supporting core research & development activities, clinical preparations, and quality and operational needs, benefitting from their leadership as they help to maximize the potential impact of Gene Coding not only on the genetic medicines industry, but on patients around the world, and the potential of SalioGens Gene Coding approach, including its use in potentially curative genetic medicines. While SalioGen believes these forward-looking statements are reasonable, undue reliance should not be placed on any such forward-looking statements, which are based on information available to us on the date of this release. These forward-looking statements are subject to various risks and uncertainties, many of which are difficult to predict, that could cause actual results to differ materially from current expectations and assumptions from those set forth or implied by any forward-looking statements. Important factors that could cause actual results to differ materially from current expectations include, among others, the ability of SalioGen to position multiple therapeutic programs for clinical development, the ability of SalioGen to continue building out its Gene Coding platform, expand the companys team, establish manufacturing and automation capabilities critical for Gene Coding and accelerate the advancement of SalioGens preclinical programs as planned, the ability of SalioGen to use its Gene Coding platform and Exact DNA Integration Technology in potentially curative genetic medicines. All forward-looking statements are based on SalioGens expectations and assumptions as of the date of this press release. Actual results may differ materially from these forward-looking statements. Except as required by law, SalioGen expressly disclaims any responsibility to update any forward-looking statement contained herein, whether as a result of new information, future events or otherwise.

Corporate Contact:Sung You, M.S., MBASalioGen Therapeuticsinvestors@saliogen.com

Media Contact:Amy Jobe, Ph.D.LifeSci Communications315-879-8192ajobe@lifescicomms.com

View post:
SalioGen Therapeutics Strengthens its Leadership Team to Advance its Gene Coding Platform - BioSpace

Posted in Genetic Medicine | Comments Off on SalioGen Therapeutics Strengthens its Leadership Team to Advance its Gene Coding Platform – BioSpace

BridgeBio Pharma Announces Positive Interim Results from a Phase 2 Trial of Infigratinib in Achondroplasia Demonstrating an Increase in Annualized…

News and research before you hear about it on CNBC and others. Claim your 1-week free trial to StreetInsider Premium here.

- At the highest dose level evaluated to date (Cohort 4, 0.128 mg/kg once daily), the mean change from baseline in annualized height velocity (AHV) was +1.52 cm/yr (p=0.02, n=11) and the responder rate was 64% in children 5 years and older1

- Infigratinib was well-tolerated with no serious adverse events (SAE) and no discontinuations due to adverse events (AE) including in Cohort 5 (dose: 0.25 mg/kg once daily) participants dosed to date, with a median duration of follow-up of 48.1 weeks; only a limited number of AEs were assessed as related to study drug and all were Grade 1, the lowest level

- Given infigratinibs profile to date, and after discussions with regulators, BridgeBio has begun dosing children in Cohort 5 (dose: 0.25 mg/kg once daily)

- Infigratinib is the only known oral product candidate currently under clinical investigation for achondroplasia, with granted and filed patent applications expiring as late as 2041

PALO ALTO, Calif., July 26, 2022 (GLOBE NEWSWIRE) -- BridgeBio Pharma, Inc. (Nasdaq: BBIO) (BridgeBio), a commercial-stage biopharmaceutical company focused on genetic diseases and cancers, today announced positive interim results from PROPEL 2, a Phase 2 trial of the investigational therapy infigratinib in children with achondroplasia. Achondroplasia (ACH) is the most common cause of disproportionate short stature. Achondroplasia impacts health and can lead to medical complications such as obstructive sleep apnea, middle ear dysfunction, kyphosis, and spinal stenosis. Achondroplasia affects approximately 55,000 people in the United States (US) and Europe, including up to 13,000 children and adolescents with open growth plates. The condition is uniformly caused by an activating mutation in fibroblast growth factor receptor 3 (FGFR3). Infigratinib is an oral small molecule that inhibits FGFR3, therefore designed to target this well-described disease at its source.

At the highest dose level evaluated to date (0.128 mg/kg once daily), mean increase in annualized height velocity (AHV) was 1.52 cm/yr over baseline (p=0.02, n=11) for all follow-up data available at time of data cut in children 5 years of age and older. In this group, 64% were responders (defined with a strict criterion of an increase 25% in AHV from baseline) and the average percent change from baseline in AHV was 60%.

Earlier cohorts in PROPEL 2 did not achieve the target efficacious exposure as suggested by our preclinical data and no dose response was observed. An increase in AHV over baseline of 0.22 cm/yr (p=0.54, n=38) was observed across these earlier combined cohorts for children 5 years of age and older.

PROPEL 2 enrolled children as young as 3 years of age in the study. Consistent with other trials in the younger age population, both AHV and height Z-scores were analyzed to help control for greater variability in growth seen by age and sex in children 3 to n=5) was observed. Across Cohort 4, the median duration of follow-up was 26.9 weeks at time of data cut.

These interim data demonstrate compelling initial proof-of-concept for an oral FGFR inhibitor in children with achondroplasia. We are encouraged by what we have seen to date, including the overall safety profile and promising initial efficacy data of infigratinib. We look forward to completing enrollment in Cohort 5 with the goal of presenting the full study results next year, said Professor Ravi Savarirayan, M.D., Ph.D., clinical geneticist and group leader of skeletal therapies research at the Murdoch Childrens Research Institute in Australia, the lead investigator for PROPEL 2.

Median follow-up across the entire study is 48.1 weeks across 62 participants included in the safety population at time of data cut. No treatment-related SAEs have been reported to date in any cohort. 90.3% of participants experienced at least one treatment-emergent adverse event (TEAE), the majority of which were Grade 1, unrelated to study drug, and consistent with a pediatric achondroplasia population. Only 9.7% of participants had a TEAE related to study drug, all of which were Grade 1, and included dyspepsia, decreased appetite, flatulence, hypercholesterolemia, hyperphosphatemia, and vitamin D decrease.

A single case of mild hyperphosphatemia (Grade 1) led to a dose reduction in a participant in Cohort 3 (dose: 0.064 mg/kg once daily), the only dose adjustment made to date in the study. Phosphorus levels for this participant only exceeded upper limit of normal by

No children have discontinued treatment as the result of an adverse event. No bone-related AEs were observed to date.

I am encouraged by this interim efficacy and safety data and thankful, as ever, to be partnered with the healthcare providers, children, and families who are making this study possible. These data, combined with our positive proof-of-concept data in LGMD2i and ADH1 earlier this year, highlight BridgeBios ability to efficiently prosecute high value programs in large areas of unmet need. We look forward to exploring the potential of infigratinib in achondroplasia and related skeletal dysplasias in the near future, said Neil Kumar, Ph.D., founder and CEO of BridgeBio.

BridgeBio, after discussions with regulatory agencies, has begun enrolling Cohort 5. Cohort 5 participants are receiving approximately twice the dose of Cohort 4 (0.25 mg/kg once daily vs 0.128 mg/kg once daily in Cohort 4). At the conclusion of the ongoing trial, BridgeBio intends to present full data at a medical conference in the first half of 2023. Additionally, BridgeBio expects to evaluate development of infigratinib in other FGFR-driven skeletal dysplasias, which affect more than 50,000 people in the US and Europe, building on this positive interim data from PROPEL 2 as well as preclinical data in hypochondroplasia presented at the Endocrine Society (ENDO) 2022 Annual Conference earlier this year.

Infigratinib is not approved in any country for the treatment of children or adults with achondroplasia. For more information on infigratinib, please see the following link.

References1Response defined as 25% increase in AHV from baseline

About BridgeBio Pharma, Inc.BridgeBio Pharma (BridgeBio) is a commercial-stage biopharmaceutical company founded to discover, create, test and deliver transformative medicines to treat patients who suffer from genetic diseases and cancers with clear genetic drivers. BridgeBios pipeline of development programs ranges from early science to advanced clinical trials. BridgeBio was founded in 2015 and its team of experienced drug discoverers, developers and innovators are committed to applying advances in genetic medicine to help patients as quickly as possible. For more information visitbridgebio.comand follow us onLinkedInandTwitter.

BridgeBio Pharma, Inc. Forward-Looking Statements

This press release contains forward-looking statements. Statements we make in this press release may include statements that are not historical facts and are considered forward-looking within the meaning of Section 27A of the Securities Act of 1933, as amended (the Securities Act), and Section 21E of the Securities Exchange Act of 1934, as amended (the Exchange Act), which are usually identified by the use of words such as anticipates, believes, estimates, expects, intends, may, plans, projects, seeks, should, will, and variations of such words or similar expressions. We intend these forward-looking statements to be covered by the safe harbor provisions for forward-looking statements contained in Section 27A of the Securities Act and Section 21E of the Exchange Act and are making this statement for purposes of complying with those safe harbor provisions. These forward-looking statements, including statements relating to the timing and success of BridgeBios Phase 2 trial of infigratinib, an investigational therapy for the treatment of children with achondroplasia, including BridgeBios ability to complete enrollment and dosing in Cohort 5, expectations, plans and prospects regarding BridgeBios regulatory approval process for infigratinib, cuts of current data being indicative of future cuts of follow-up data, interim results being predictive of final results, interim efficacy and safety data being predictive of final efficacy and safety data, the potential for infigratinib to be the first oral product for the treatment of achondroplasia, BridgeBios ability to secure intellectual property protection for infigratinib and the duration of such protection, the potential patient population that infigratinib, if approved, could address in the United States and Europe, BridgeBios ability to efficiently prosecute high value programs in large areas of unmet need, BridgeBios ability to develop infigratinib in other FGFR-driven skeletal dysplasias, and the success of BridgeBios continuing discussions with regulatory agencies regarding dosing and trial design, reflect our current views about our plans, intentions, expectations, strategies and prospects, which are based on the information currently available to us and on assumptions we have made. Although we believe that our plans, intentions, expectations, strategies and prospects as reflected in or suggested by those forward-looking statements are reasonable, we can give no assurance that the plans, intentions, expectations or strategies will be attained or achieved. Furthermore, actual results may differ materially from those described in the forward-looking statements and will be affected by a number of risks, uncertainties and assumptions, including, but not limited to, BridgeBios ability to complete the Phase 2 trial of infigratinib, including its ability to complete enrollment and dosing in Cohort 5, the success of cuts of current data not being indicative of future cuts of follow-up data, interim results not being predictive of final results, interim efficacy and safety data not being predictive of final efficacy and safety data, the potential for infigratinib to be the first oral product for the treatment of achondroplasia, BridgeBios ability to obtain intellectual property protection for infigratinib and the duration of any such protection, BridgeBios ability to advance infigratinib in clinical development according to its plans and discussions with regulatory agencies, and potential adverse impacts due to the global COVID-19 pandemic such as delays in regulatory review, manufacturing and clinical trials, supply chain interruptions, adverse effects on healthcare systems and disruption of the global economy; as well as those set forth in the Risk Factors section of BridgeBios most recent Annual Report on Form 10-K filed with the U.S. Securities and Exchange Commission (SEC) and in subsequent SEC filings, which are available on the SECs website atwww.sec.gov. Moreover, BridgeBio operates in a very competitive and rapidly changing environment in which new risks emerge from time to time. Except as required by applicable law, we assume no obligation to update publicly any forward-looking statements, whether as a result of new information, future events or otherwise.

BridgeBio Contact:Grace Rauh[emailprotected](917) 232-5478

Here is the original post:
BridgeBio Pharma Announces Positive Interim Results from a Phase 2 Trial of Infigratinib in Achondroplasia Demonstrating an Increase in Annualized...

Posted in Genetic Medicine | Comments Off on BridgeBio Pharma Announces Positive Interim Results from a Phase 2 Trial of Infigratinib in Achondroplasia Demonstrating an Increase in Annualized…