Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Diagnostics (Basel) ; 13(11)2023 May 28.
Article in English | MEDLINE | ID: mdl-37296738

ABSTRACT

COVID-19, continually developing and raising increasingly significant issues, has impacted human health and caused countless deaths. It is an infectious disease with a high incidence and mortality rate. The spread of the disease is also a significant threat to human health, especially in the developing world. This study suggests a method called shuffle shepherd optimization-based generalized deep convolutional fuzzy network (SSO-GDCFN) to diagnose the COVID-19 disease state, types, and recovered categories. The results show that the accuracy of the proposed method is as high as 99.99%; similarly, precision is 99.98%; sensitivity/recall is 100%; specificity is 95%; kappa is 0.965%; AUC is 0.88%; and MSE is less than 0.07% as well as 25 s. Moreover, the performance of the suggested method has been confirmed by comparison of the simulation results from the proposed approach with those from several traditional techniques. The experimental findings demonstrate strong performance and high accuracy for categorizing COVID-19 stages with minimal reclassifications over the conventional methods.

2.
Mol Metab ; 66: 101624, 2022 12.
Article in English | MEDLINE | ID: mdl-36341906

ABSTRACT

OBJECTIVE: Lifelong insulin replacement remains the mainstay of type 1 diabetes treatment. Genetic FoxO1 ablation promotes enteroendocrine cell (EECs) conversion into glucose-responsive ß-like cells. Here, we tested whether chemical FoxO1 inhibitors can generate ß-like gut cells. METHODS: We used Ngn3-or Villin-driven FoxO1 ablation to capture the distinctive developmental effects of FoxO1 on EEC pool. We combined FoxO1 ablation with Notch inhibition to enhance the expansion of EEC pool. We tested the ability of an orally available small molecule of FoxO1 inhibitor, Cpd10, to phenocopy genetic ablation of FoxO1. We evaluated the therapeutic impact of genetic ablation or chemical inhibition of FoxO1 on insulin-deficient diabetes in Ins2Akita/+ mice. RESULTS: Pan-intestinal epithelial FoxO1 ablation expanded the EEC pool, induced ß-like cells, and improved glucose tolerance in Ins2Akita/+ mice. This genetic effect was phenocopied by Cpd10. Cpd10 induced ß-like cells that released insulin in response to glucose in gut organoids, and this effect was enhanced by the Notch inhibitor, DBZ. In Ins2Akita/+ mice, a five-day course of either Cpd10 or DBZ induced intestinal insulin-immunoreactive ß-like cells, lowered glycemia, and increased plasma insulin levels without apparent adverse effects. CONCLUSION: These results provide proof of principle of gut cell conversion into ß-like cells by a small molecule FoxO1 inhibitor, paving the way for clinical applications.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Animals , Mice , Enteroendocrine Cells , Forkhead Box Protein O1/genetics , Glucose/pharmacology , Insulin/genetics , Organoids , Receptors, Notch/antagonists & inhibitors
3.
J Clin Invest ; 132(24)2022 12 15.
Article in English | MEDLINE | ID: mdl-36282594

ABSTRACT

As a highly regenerative organ, the intestine is a promising source for cellular reprogramming for replacing lost pancreatic ß cells in diabetes. Gut enterochromaffin cells can be converted to insulin-producing cells by forkhead box O1 (FoxO1) ablation, but their numbers are limited. In this study, we report that insulin-immunoreactive cells with Paneth/goblet cell features are present in human fetal intestine. Accordingly, lineage-tracing experiments show that, upon genetic or pharmacologic FoxO1 ablation, the Paneth/goblet lineage can also undergo conversion to the insulin lineage. We designed a screening platform in gut organoids to accurately quantitate ß-like cell reprogramming and fine-tune a combination treatment to increase the efficiency of the conversion process in mice and human adult intestinal organoids. We identified a triple blockade of FOXO1, Notch, and TGF-ß that, when tested in insulin-deficient streptozotocin (STZ) or NOD diabetic animals, resulted in near normalization of glucose levels, associated with the generation of intestinal insulin-producing cells. The findings illustrate a therapeutic approach for replacing insulin treatment in diabetes.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Humans , Mice , Animals , Forkhead Box Protein O1/genetics , Forkhead Transcription Factors/genetics , Mice, Inbred NOD , Insulin/genetics
4.
Mol Metab ; 66: 101618, 2022 12.
Article in English | MEDLINE | ID: mdl-36283677

ABSTRACT

OBJECTIVES: Insulin treatment remains the sole effective intervention for Type 1 Diabetes. Here, we investigated the therapeutic potential of converting intestinal epithelial cells to insulin-producing, glucose-responsive ß-like cells by targeted inhibition of FOXO1. We have previously shown that this can be achieved by genetic ablation in gut Neurogenin3 progenitors, adenoviral or shRNA-mediated inhibition in human gut organoids, and chemical inhibition in Akita mice, a model of insulin-deficient diabetes. METHODS: We profiled two novel FOXO1 inhibitors in reporter gene assays, and hepatocyte gene expression studies, and in vivo pyruvate tolerance test (PTT) for their activity and specificity. We evaluated their glucose-lowering effect in mice rendered insulin-deficient by administration of streptozotocin. RESULTS: We provide evidence that two novel FOXO1 inhibitors, FBT432 and FBT374 have glucose-lowering and gut ß-like cell-inducing properties in mice. FBT432 is also highly effective in combination with a Notch inhibitor in this model. CONCLUSION: The data add to a growing body of evidence suggesting that FOXO1 inhibition be pursued as an alternative treatment to insulin administration in diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Forkhead Box Protein O1 , Animals , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Glucose/metabolism , Insulin/metabolism , Streptozocin
5.
Preprint in English | bioRxiv | ID: ppbiorxiv-511397

ABSTRACT

COVID-19 pandemic keeps pressing onward and effective treatment option against it is still far-off. Since the onslaught in 2020, 13 different variants of SARS-CoV-2 have been surfaced including 05 different variants of concern. Success in faster pandemic handling in the future largely depends on reinforcing therapeutics along with vaccines. As a part of RNAi therapeutics, here we developed a computational approach for predicting siRNAs, which are presumed to be intrinsically active against two crucial mRNAs of SARS-CoV-2, the RNA-dependent RNA polymerase (RdRp), and the nucleocapsid phosphoprotein gene (N gene). Sequence conservancy among the alpha, beta, gamma, and delta variants of SARS-CoV-2 was integrated in the analyses that warrants the potential of these siRNAs against multiple variants. We preliminary found 13 RdRP-targeting and 7 N gene-targeting siRNAs using the siDirect V.2.0. These siRNAs were subsequently filtered through different parameters at optimum condition including macromolecular docking studies. As a result, we selected 4 siRNAs against the RdRP and 3 siRNAs against the N-gene as RNAi candidates. Development of these potential siRNA therapeutics can significantly synergize COVID-19 mitigation by lessening the efforts, furthermore, can lay a rudimentary base for the in silico design of RNAi therapeutics for future emergencies.

6.
Mol Ther ; 29(10): 3042-3058, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34332145

ABSTRACT

Reprogramming non-cardiomyocytes (non-CMs) into cardiomyocyte (CM)-like cells is a promising strategy for cardiac regeneration in conditions such as ischemic heart disease. Here, we used a modified mRNA (modRNA) gene delivery platform to deliver a cocktail, termed 7G-modRNA, of four cardiac-reprogramming genes-Gata4 (G), Mef2c (M), Tbx5 (T), and Hand2 (H)-together with three reprogramming-helper genes-dominant-negative (DN)-TGFß, DN-Wnt8a, and acid ceramidase (AC)-to induce CM-like cells. We showed that 7G-modRNA reprogrammed 57% of CM-like cells in vitro. Through a lineage-tracing model, we determined that delivering the 7G-modRNA cocktail at the time of myocardial infarction reprogrammed ∼25% of CM-like cells in the scar area and significantly improved cardiac function, scar size, long-term survival, and capillary density. Mechanistically, we determined that while 7G-modRNA cannot create de novo beating CMs in vitro or in vivo, it can significantly upregulate pro-angiogenic mesenchymal stromal cells markers and transcription factors. We also demonstrated that our 7G-modRNA cocktail leads to neovascularization in ischemic-limb injury, indicating CM-like cells importance in other organs besides the heart. modRNA is currently being used around the globe for vaccination against COVID-19, and this study proves this is a safe, highly efficient gene delivery approach with therapeutic potential to treat ischemic diseases.


Subject(s)
Cellular Reprogramming/genetics , Genetic Therapy/methods , Ischemia/therapy , Muscle, Skeletal/blood supply , Myocardial Infarction/therapy , Neovascularization, Physiologic/genetics , Regeneration/genetics , Transfection/methods , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Knockout, ApoE , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics
7.
Adv Sci (Weinh) ; 8(10): 2004661, 2021 05.
Article in English | MEDLINE | ID: mdl-34026458

ABSTRACT

Heart failure (HF) remains a major cause of morbidity and mortality worldwide. One of the risk factors for HF is cardiac hypertrophy (CH), which is frequently accompanied by cardiac fibrosis (CF). CH and CF are controlled by master regulators mTORC1 and TGF-ß, respectively. Type-2-phosphatidylinositol-5-phosphate-4-kinase-gamma (Pip4k2c) is a known mTORC1 regulator. It is shown that Pip4k2c is significantly downregulated in the hearts of CH and HF patients as compared to non-injured hearts. The role of Pip4k2c in the heart during development and disease is unknown. It is shown that deleting Pip4k2c does not affect normal embryonic cardiac development; however, three weeks after TAC, adult Pip4k2c-/- mice has higher rates of CH, CF, and sudden death than wild-type mice. In a gain-of-function study using a TAC mouse model, Pip4k2c is transiently upregulated using a modified mRNA (modRNA) gene delivery platform, which significantly improve heart function, reverse CH and CF, and lead to increased survival. Mechanistically, it is shown that Pip4k2c inhibits TGFß1 via its N-terminal motif, Pip5k1α, phospho-AKT 1/2/3, and phospho-Smad3. In sum, loss-and-gain-of-function studies in a TAC mouse model are used to identify Pip4k2c as a potential therapeutic target for CF, CH, and HF, for which modRNA is a highly translatable gene therapy approach.


Subject(s)
Cardiomegaly/complications , Fibrosis/prevention & control , Heart Failure/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/physiology , RNA, Messenger/genetics , Adult , Aged , Animals , Cellular Reprogramming , Disease Models, Animal , Female , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phosphotransferases (Alcohol Group Acceptor)/administration & dosage , RNA, Messenger/administration & dosage , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling
8.
Methods Mol Biol ; 2158: 281-294, 2021.
Article in English | MEDLINE | ID: mdl-32857381

ABSTRACT

Modified mRNA (modRNA) is a promising new gene therapy approach that has safely and effectively delivered genes into different tissues, including the heart. Current efforts to use DNA-based or viral gene therapy to induce cardiac regeneration postmyocardial infarction (MI) or in heart failure (HF) have encountered key challenges, e.g., genome integration and delayed and noncontrolled expression. By contrast, modRNA is a transient, safe, non-immunogenic, and controlled gene delivery method that is not integrated into the genome. For most therapeutic applications, especially in regenerative medicine, the ability to deliver genes to the heart transiently and with control is vital for achieving therapeutic effect. Additionally, modRNA synthesis is comparatively simple and inexpensive compared to other gene delivery methods (e.g., protein), though a simple, clear in vitro transcription (IVT) protocol for synthesizing modRNA is needed for it to be more widely used. Here, we describe a simple and improved step-by-step IVT protocol to synthesize modRNA for in vitro or in vivo applications.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Myocardial Infarction/therapy , Myocardium/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/chemistry , Regenerative Medicine , Transcription, Genetic , Animals , Mice , Myocardial Infarction/genetics , RNA, Messenger/genetics
9.
J Vis Exp ; (160)2020 06 11.
Article in English | MEDLINE | ID: mdl-32597835

ABSTRACT

Myocardial infarction (MI) is a leading cause of morbidity and mortality in the Western world. In the past decade, gene therapy has become a promising treatment option for heart disease, owing to its efficiency and exceptional therapeutic effects. In an effort to repair the damaged tissue post-MI, various studies have employed DNA-based or viral gene therapy but have faced considerable hurdles due to the poor and uncontrolled expression of the delivered genes, edema, arrhythmia, and cardiac hypertrophy. Synthetic modified mRNA (modRNA) presents a novel gene therapy approach that offers high, transient, safe, nonimmunogenic, and controlled mRNA delivery to the heart tissue without any risk of genomic integration. Due to these remarkable characteristics combined with its bell-shaped pharmacokinetics in the heart, modRNA has become an attractive approach for the treatment of heart disease. However, to increase its effectiveness in vivo, a consistent and reliable delivery method needs to be followed. Hence, to maximize modRNA delivery efficiency and yield consistency in modRNA use for in vivo applications, an optimized method of preparation and delivery of modRNA intracardiac injection in a mouse MI model is presented. This protocol will make modRNA delivery more accessible for basic and translational research.


Subject(s)
Gene Transfer Techniques , Myocardial Infarction/genetics , Myocardial Infarction/therapy , RNA, Messenger/administration & dosage , RNA, Messenger/therapeutic use , Animals , Disease Models, Animal , Genetic Therapy/methods , Injections , Integrases/metabolism , Ligation , Luciferases/metabolism , Mice , Myocardial Infarction/surgery , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
10.
Mol Ther Methods Clin Dev ; 17: 622-633, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32300609

ABSTRACT

Modified mRNA (modRNA) is a gene-delivery platform for transiently introducing a single gene or several genes of interest to different cell types and tissues. modRNA is considered to be a safe vector for gene transfer, as it negligibly activates the innate immune system and does not compromise the genome integrity. The use of modRNA in basic and translational science is rising, due to the clinical potential of modRNA. We are currently using modRNA to induce cardiac regeneration post-ischemic injury. Major obstacles in using modRNA for cardiac ischemic disease include the need for the direct and single administration of modRNA to the heart and the inefficient translation of modRNA due to its short half-life. Modulation of the 5' untranslated region (5' UTR) to enhance translation efficiency in ischemic cardiac disease has great value, as it can reduce the amount of modRNA needed per delivery and will achieve higher and longer protein production post-single delivery. Here, we identified that 5' UTR, from the fatty acid metabolism gene carboxylesterase 1D (Ces1d), enhanced the translation of firefly luciferase (Luc) modRNA by 2-fold in the heart post-myocardial infarction (MI). Moreover, we identified, in the Ces1d, a specific RNA element (element D) that is responsible for the improvement of modRNA translation and leads to a 2.5-fold translation increment over Luc modRNA carrying artificial 5' UTR, post-MI. Importantly, we were able to show that 5' UTR Ces1d also enhances modRNA translation in the liver, but not in the kidney, post-ischemic injury, indicating that Ces1d 5' UTR and element D may play a wider role in translation of protein under an ischemic condition.

11.
Circulation ; 141(11): 916-930, 2020 03 17.
Article in English | MEDLINE | ID: mdl-31992066

ABSTRACT

BACKGROUND: Sphingolipids have recently emerged as a biomarker of recurrence and mortality after myocardial infarction (MI). The increased ceramide levels in mammalian heart tissues during acute MI, as demonstrated by several groups, is associated with higher cell death rates in the left ventricle and deteriorated cardiac function. Ceramidase, the only enzyme known to hydrolyze proapoptotic ceramide, generates sphingosine, which is then phosphorylated by sphingosine kinase to produce the prosurvival molecule sphingosine-1-phosphate. We hypothesized that Acid Ceramidase (AC) overexpression would counteract the negative effects of elevated ceramide and promote cell survival, thereby providing cardioprotection after MI. METHODS: We performed transcriptomic, sphingolipid, and protein analyses to evaluate sphingolipid metabolism and signaling post-MI. We investigated the effect of altering ceramide metabolism through a loss (chemical inhibitors) or gain (modified mRNA [modRNA]) of AC function post hypoxia or MI. RESULTS: We found that several genes involved in de novo ceramide synthesis were upregulated and that ceramide (C16, C20, C20:1, and C24) levels had significantly increased 24 hours after MI. AC inhibition after hypoxia or MI resulted in reduced AC activity and increased cell death. By contrast, enhancing AC activity via AC modRNA treatment increased cell survival after hypoxia or MI. AC modRNA-treated mice had significantly better heart function, longer survival, and smaller scar size than control mice 28 days post-MI. We attributed the improvement in heart function post-MI after AC modRNA delivery to decreased ceramide levels, lower cell death rates, and changes in the composition of the immune cell population in the left ventricle manifested by lowered abundance of proinflammatory detrimental neutrophils. CONCLUSIONS: Our findings suggest that transiently altering sphingolipid metabolism through AC overexpression is sufficient and necessary to induce cardioprotection post-MI, thereby highlighting the therapeutic potential of AC modRNA in ischemic heart disease.


Subject(s)
Acid Ceramidase/physiology , Genetic Therapy , Hypoxia/metabolism , Myocardial Infarction/metabolism , RNA, Messenger/therapeutic use , Sphingolipids/metabolism , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/genetics , Animals , Animals, Newborn , Apoptosis , Ceramides/metabolism , Cicatrix/pathology , Embryoid Bodies , Enzyme Induction , Female , Humans , Hypoxia/etiology , Hypoxia/pathology , Induced Pluripotent Stem Cells/metabolism , Inflammation , Male , Mice , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Transfection , Up-Regulation
12.
Mol Ther Methods Clin Dev ; 14: 300-305, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31508439

ABSTRACT

Synthetic modified RNA (modRNA) is a novel vector for gene transfer to the heart and other organs. modRNA can mediate strong, transient protein expression with minimal induction of the innate immune response and risk for genome integration. modRNA is already being used in several human clinical trials, and its use in basic and translational science is growing. Due to the complexity of preparing modRNA and the high cost of its reagents, there is a need for an improved, cost-efficient protocol to make modRNA. Here we show that changing the ratio between anti-reverse cap analog (ARCA) and N1-methyl-pseudouridine (N1mΨ), favoring ARCA over N1mΨ, significantly increases the yield per reaction, improves modRNA translation, and reduces its immunogenicity in vitro. This protocol will make modRNA preparation more accessible and financially affordable for basic and translational research.

13.
PLoS Negl Trop Dis ; 13(1): e0007124, 2019 01.
Article in English | MEDLINE | ID: mdl-30703097

ABSTRACT

BACKGROUND: Cholera remains a substantial health burden in Asia and Africa particularly in resource poor settings. The standard procedures to identify the etiological organism V. cholerae are isolation from microbiological culture from stool as well as Polymerase Chain Reaction (PCR). Both the processes are highly lab oriented, labor extensive, time consuming, and expensive. In an effort to control for outbreaks and epidemics; an effective, convenient, quick and relatively less expensive detection method is imperative, without compromising the sensitivity and specificity that exists at present. The objective of this component of the study was to evaluate the effectiveness of a locally produced rapid diagnostic test (RDT) for cholera diagnosis. METHODS: In Bangladesh, nationwide cholera surveillance is ongoing in 22 hospitals covering all 8 divisions of the country since June, 2016. In the surveillance, stool samples have been collected from patients presenting to hospitals with acute watery diarrhea. Crystal VCTM (Span diagnostics, India) and Cholkit (locally produced RDT) have been used to detect V. cholerae from stool samples. Samples have also been sent to the main laboratory at icddr,b where the culture based isolation is routinely performed. All the tests were carried out for both direct and enriched stool samples. RDT sensitivity and specificity were calculated using stool culture as the gold standard. RESULTS: A total of 7720 samples were tested. Among these, 5865 samples were solely tested with Crystal VC and 1355 samples with Cholkit whereas 381 samples were tested with both the RDTs. In comparison with culture, direct testing with Crystal VC showed a sensitivity of 72% (95% CI: 50.6% to 87.9%) and specificity of 86.8% (95% CI: 82.8% to 90.1%). After enrichment the sensitivity and specificity was 68% (95% CI: 46.5% to 85.1%) and 97.5% (95% CI: 95.3% to 98.8%) respectively. The direct Cholkit test showed sensitivity of 76% (95% CI: 54.9% to 90.6%) and specificity of 90.2% (95% CI: 86.6% to 93.1%). CONCLUSION: This evaluation has demonstrated that the sensitivity and specificity of Cholkit is similar to the commercially available test, Crystal VC when used in field settings for detecting V. cholerae from stool specimens. The findings from this study suggest that the Cholkit could be a possible alternative for cholera endemic regions where V. cholerae O1 is the major causative organism causing cholera.


Subject(s)
Cholera/diagnosis , Vibrio cholerae/isolation & purification , Adolescent , Antibodies, Bacterial/analysis , Antibodies, Bacterial/immunology , Bangladesh , Child , Child, Preschool , Diarrhea , Early Diagnosis , Feces/microbiology , Female , Humans , Male , O Antigens/analysis , O Antigens/immunology , Reagent Kits, Diagnostic , Sensitivity and Specificity , Serotyping
14.
Circulation ; 138(25): 2919-2930, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566018

ABSTRACT

BACKGROUND: For more than a decade, Sca-1+ cells within the mouse heart have been widely recognized as a stem cell population with multipotency that can give rise to cardiomyocytes, endothelial cells, and smooth muscle cells in vitro and after cardiac grafting. However, the developmental origin and authentic nature of these cells remain elusive. METHODS: Here, we used a series of high-fidelity genetic mouse models to characterize the identity and regenerative potential of cardiac resident Sca-1+ cells. RESULTS: With these novel genetic tools, we found that Sca-1 does not label cardiac precursor cells during early embryonic heart formation. Postnatal cardiac resident Sca-1+ cells are in fact a pure endothelial cell population. They retain endothelial properties and exhibit minimal cardiomyogenic potential during development, normal aging and upon ischemic injury. CONCLUSIONS: Our study provides definitive insights into the nature of cardiac resident Sca-1+ cells. The observations challenge the current dogma that cardiac resident Sca-1+ cells are intrinsic stem cells for myocardial development, renewal, and repair, and suggest that the mechanisms of transplanted Sca-1+ cells in heart repair need to be reassessed.


Subject(s)
Adult Stem Cells/physiology , Antigens, Ly/metabolism , Endothelial Cells/physiology , Heart/embryology , Membrane Proteins/metabolism , Myocytes, Cardiac/physiology , Animals , Antigens, Ly/genetics , Cell Differentiation , Cell Lineage , Cell Self Renewal , Cells, Cultured , Embryonic Development , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Models, Animal , Regeneration , Stem Cell Transplantation , Wound Healing
15.
J Biol Chem ; 293(24): 9162-9175, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29735531

ABSTRACT

Chondrocyte hypertrophy is the terminal step in chondrocyte differentiation and is crucial for endochondral bone formation. How signaling pathways regulate chondrocyte hypertrophic differentiation remains incompletely understood. In this study, using a Tbx18:Cre (Tbx18Cre/+) gene-deletion approach, we selectively deleted the gene for the signaling protein SMAD family member 4 (Smad4f/f ) in the limbs of mice. We found that the Smad4-deficient mice develop a prominent shortened limb, with decreased expression of chondrocyte differentiation markers, including Col2a1 and Acan, in the humerus at mid-to-late gestation. The most striking defects in these mice were the absence of stylopod elements and failure of chondrocyte hypertrophy in the humerus. Moreover, expression levels of the chondrocyte hypertrophy-related markers Col10a1 and Panx3 were significantly decreased. Of note, we also observed that the expression of runt-related transcription factor 2 (Runx2), a critical mediator of chondrocyte hypertrophy, was also down-regulated in Smad4-deficient limbs. To determine how the skeletal defects arose in the mouse mutants, we performed RNA-Seq with ChIP-Seq analyses and found that Smad4 directly binds to regulatory elements in the Runx2 promoter. Our results suggest a new mechanism whereby Smad4 controls chondrocyte hypertrophy by up-regulating Runx2 expression during skeletal development. The regulatory mechanism involving Smad4-mediated Runx2 activation uncovered here provides critical insights into bone development and pathogenesis of chondrodysplasia.


Subject(s)
Bone Development , Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Smad4 Protein/genetics , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Core Binding Factor Alpha 1 Subunit/metabolism , Hypertrophy/genetics , Hypertrophy/metabolism , Hypertrophy/pathology , Mice , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Smad4 Protein/metabolism
16.
PLoS Negl Trop Dis ; 12(3): e0006286, 2018 03.
Article in English | MEDLINE | ID: mdl-29538377

ABSTRACT

Recognizing cholera cases early, especially in the initial phase of an outbreak and in areas where cholera has not previously circulated, is a high public health priority. Laboratory capacity in such settings is often limited. To address this, we have developed a rapid diagnostic test (RDT) termed Cholkit that is based on an immunochromatographic lateral flow assay for the diagnosis of cholera cases using stool. Cholkit contains a monoclonal antibody (ICL-33) to the O-specific polysaccharide (OSP) component of V. cholerae O1 lipopolysaccharide, and recognizes both Inaba and Ogawa serotypes. We tested the Cholkit dipstick using fresh stool specimens of 76 adults and children presenting with acute watery diarrhea at the icddr,b hospital in Dhaka, Bangladesh. We compared Cholkit's performance with those of microbial culture, PCR (targeting the rfb and ctxA genes of V. cholerae) and the commercially available RDT, Crystal VC (Span Diagnostics; Surat, India). We found that all stool specimens with a positive culture for V. cholerae O1 (n = 19) were positive by Cholkit as well as Crystal VC. We then used Bayesian latent class modeling to estimate the sensitivity and specificity of each diagnostic assay. The sensitivity of Cholkit, microbiological culture, PCR and Crystal VC was 98% (95% CI: 88-100), 71% (95% CI: 59-81), 74% (95% CI: 59-86) and 98% (95% CI: 88-100), respectively. The specificity for V. cholerae O1 was 97% (95% CI: 89-100), 100%, 97% (95% CI: 93-99) and 98% (95% CI: 92-100), respectively. Of note, two Crystal VC dipsticks were positive for V. cholerae O139 but negative by culture and PCR in this area without known circulating epidemic V. cholerae O139. In conclusion, the Cholkit dipstick is simple to use, requires no dedicated laboratory capacity, and has a sensitivity and specificity for V. cholerae O1 of 98% and 97%, respectively. Cholkit warrants further evaluation in other settings.


Subject(s)
Bacteriological Techniques , Cholera/diagnosis , Diarrhea/microbiology , Feces/microbiology , O Antigens/analysis , Vibrio cholerae O1/isolation & purification , Acute Disease , Adolescent , Adult , Bangladesh/epidemiology , Child , Child, Preschool , Cholera/epidemiology , Cholera/immunology , Cholera/microbiology , Diarrhea/epidemiology , Disease Outbreaks , Female , Hospitalization , Humans , Infant , Male , Middle Aged , Polymerase Chain Reaction , Reagent Kits, Diagnostic , Sensitivity and Specificity , Vibrio cholerae O1/genetics , Vibrio cholerae O1/immunology , Young Adult
17.
Indian J Dent Res ; 28(5): 545-548, 2017.
Article in English | MEDLINE | ID: mdl-29072218

ABSTRACT

Context (Background): Lips prints are unique and are a tool for personal identification. AIMS: Indian population can be divided into different ethno-racial groups. In this study, we aimed at finding the most and the least prevalent lip print patterns in these groups and also to observe any similarities or differences that may exist in these groups in terms of lip print patterns. SETTINGS AND DESIGN: Lip prints in 755 individuals categorized into different ethno-racial groups were studied. MATERIALS AND METHODS: Brown- and pink-colored lipsticks, cellophane tape, and magnifying lens were used to record and study the lip prints. RESULTS: Among all the three ethno-racial groups, Type I was the most prevalent lip print pattern observed. The least prevalent lip print pattern in all the three groups was Type IV. Inference/Conclusion: Lip prints hold potential as supplementary tools for identification where they can be recorded with ease. The observation and classification of lip print patterns in different ethno-racial groups not only provide some useful data but also open a new window to a field that can contribute extensively to criminal investigation and identification.


Subject(s)
Ethnicity , Lip/anatomy & histology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , India , Infant , Male , Middle Aged
18.
Mol Ther ; 25(6): 1306-1315, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28389322

ABSTRACT

Modified mRNA (modRNA) is a new technology in the field of somatic gene transfer that has been used for the delivery of genes into different tissues, including the heart. Our group and others have shown that modRNAs injected into the heart are robustly translated into the encoded protein and can potentially improve outcome in heart injury models. However, the optimal compositions of the modRNA and the reagents necessary to achieve optimal expression in the heart have not been characterized yet. In this study, our aim was to elucidate those parameters by testing different nucleotide modifications, modRNA doses, and transfection reagents both in vitro and in vivo in cardiac cells and tissue. Our results indicate that optimal cardiac delivery of modRNA is with N1-Methylpseudouridine-5'-Triphosphate nucleotide modification and achieved using 0.013 µg modRNA/mm2/500 cardiomyocytes (CMs) transfected with positively charged transfection reagent in vitro and 100 µg/mouse heart (1.6 µg modRNA/µL in 60 µL total) sucrose-citrate buffer in vivo. We have optimized the conditions for cardiac delivery of modRNA in vitro and in vivo. Using the described methods and conditions may allow for successful gene delivery using modRNA in various models of cardiovascular disease.


Subject(s)
Gene Transfer Techniques , Myocardium/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Animals , Biomarkers , Cells, Cultured , Female , Gene Expression , Genes, Reporter , Humans , Male , Mice , Molecular Imaging , Protein Biosynthesis , RNA, Messenger/chemistry , Rats , Transfection
19.
Methods Mol Biol ; 1521: 127-138, 2017.
Article in English | MEDLINE | ID: mdl-27910045

ABSTRACT

Cardiac gene therapy shows tremendous promise in combating the growing problem of heart disease. Modified mRNA (modRNA) is a novel gene delivery system used in vitro or in vivo to achieve transient expression of therapeutic proteins in a heterogeneous population of cells. Incorporation of specific modified nucleosides enables modRNA to be translated efficiently without triggering antiviral and innate immune responses. ModRNA has been shown to be effective at delivering short-term robust gene expression to the heart and its use in the field of cardiac gene therapy is expanding. Here, we describe a stepwise protocol for the synthesis of modRNA for in vivo myocardial delivery.


Subject(s)
Gene Transfer Techniques , Myocardium/metabolism , RNA, Messenger/biosynthesis , Animals , Chemical Precipitation , Mice , Plasmids/genetics , Polymerase Chain Reaction , Quality Control , Templates, Genetic , Transcription, Genetic
20.
Circulation ; 135(1): 59-72, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27803039

ABSTRACT

BACKGROUND: Epicardial adipose tissue volume and coronary artery disease are strongly associated, even after accounting for overall body mass. Despite its pathophysiological significance, the origin and paracrine signaling pathways that regulate epicardial adipose tissue's formation and expansion are unclear. METHODS: We used a novel modified mRNA-based screening approach to probe the effect of individual paracrine factors on epicardial progenitors in the adult heart. RESULTS: Using 2 independent lineage-tracing strategies in murine models, we show that cells originating from the Wt1+ mesothelial lineage, which includes epicardial cells, differentiate into epicardial adipose tissue after myocardial infarction. This differentiation process required Wt1 expression in this lineage and was stimulated by insulin-like growth factor 1 receptor (IGF1R) activation. IGF1R inhibition within this lineage significantly reduced its adipogenic differentiation in the context of exogenous, IGF1-modified mRNA stimulation. Moreover, IGF1R inhibition significantly reduced Wt1 lineage cell differentiation into adipocytes after myocardial infarction. CONCLUSIONS: Our results establish IGF1R signaling as a key pathway that governs epicardial adipose tissue formation in the context of myocardial injury by redirecting the fate of Wt1+ lineage cells. Our study also demonstrates the power of modified mRNA -based paracrine factor library screening to dissect signaling pathways that govern progenitor cell activity in homeostasis and disease.


Subject(s)
Adipocytes/metabolism , Mesenchymal Stem Cells/cytology , Myocardial Infarction/pathology , Pericardium/cytology , Receptor, IGF Type 1/metabolism , Adipocytes/cytology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Disease Models, Animal , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Myocardial Infarction/metabolism , Paracrine Communication , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/genetics , Repressor Proteins/metabolism , Signal Transduction , WT1 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...