Adsense

Tracking a Serial Killer: Ebola virus mutating rapidly as it spreads.

Why we need to terminate Ebola 2014 before the virus learns too much about us.

Biochemistry and Molecular Biology Slide 2

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Biochemistry and Molecular Biology Slide 3

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Biochemistry and Molecular Biology Slide 4

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Biochemistry and Molecular Biology Slide 5

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Biochemistry and Molecular Biology Slide 6

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Biochemistry and Molecular Biology Slide 7

This theme is Bloggerized by Dr. Shreekrishna Maharjan.

Monday, September 29, 2014

The Nutshell: FDA OKs siRNA Ebola Drug

FDA OKs siRNA Ebola Drug

The US Food and Drug Administration gives the green light to deploy an experimental short interfering RNA treatment against Ebola.

By | September 23, 2014
WIKIMEDIA, RICHARD ROBINSON
Although TKM-Ebola, a short interfering RNA (siRNA) therapy to treat Ebola infection, has not been approved for use in humans, the US Food and Drug Administration (FDA) is allowing for “compassionate use” of the medication. In other words, even though a drug is not fully vetted, the dire circumstances of the patients justify risky measures. Tekmira Pharmaceutical, the drug’s manufacturer, announced in a press release yesterday (September 22) that the FDA and Health Canada will “allow the use of our investigational therapeutic in more patients.”
 
Already, Tekmira has provided the medication to several Ebola patients “and the repeat infusions have been well tolerated,” according to the statement. “However, it must be kept in mind that any uses of the product under expanded access, does not constitute controlled clinical trials.”
 
David Kroll at Forbes pointed out that the successful administration of TKM-Ebola in monkeys saved the animals from a strain of the virus that circulated in 1995. “On one hand, this shows that TKM-Ebola could be effective against a strain separated by almost 20 years, but is no guarantee that it will against the currently-circulating strain,” he wrote.
 
USA Today reported that one of the patients who has taken TKM-Ebola is Richard Sacra, a US physician who became infected in Liberia and is now being treated in Nebraska. Sacra also received a blood transfusion from another doctor who survived the infection after taking ZMapp, an experimental monoclonal antibody therapy. According to the news report, “While it’s too early to know if TKM-Ebola will work on more patients, [TKM-Ebola researcher Thomas] Geisbert said he’s encouraged [that] Sacra was able to take the drug safely. That’s no small feat, he said. Experimental drugs with unknown effects are usually tested in healthy people. Sacra was acutely ill when he received the drug.”
 
Forbes’s Kroll cautioned that given the limited supply of medication Tekmira has available to distribute, data from patients given compassionate use may not be sufficient to offer reliable insight into the effectiveness of the drug.
http://www.the-scientist.com/?articles.view/articleNo/41081/title/FDA-OKs-siRNA-Ebola-Drug/

Latest News: How did the 'Berlin patient' rid himself of HIV?



Eric Risberg/AP Photo
 
Timothy Ray Brown, known by many researchers as "the Berlin patient," is the only person to have been cured of an HIV infection.

How did the 'Berlin patient' rid himself of HIV?

Researchers are closer to unraveling the mystery of how Timothy Ray Brown, the only human cured of HIV, defeated the virus, according to a new study. Although the work doesn’t provide a definitive answer, it rules out one possible explanation.
 
Brown remains one of the most studied cases in the HIV epidemic’s history. In 2006, after living with the virus for 11 years and controlling his infection with antiretroviral drugs (ARVs), he learned that he had developed acute myeloid leukemia. (The leukemia has no known relationship to HIV infection or treatment.) Chemotherapy failed, and the next year Brown, an American then living in Berlin, received the first of two bone marrow transplants—a common treatment for this cancer—and ditched his ARVs. When HIV-infected people stop taking ARVs, levels of HIV typically skyrocket within weeks. Yet researchers scouring Brown’s blood over the past 7 years have found only traces of the viral genetic material, none of which can replicate.
 
Today, researchers point to three different factors that could independently or in combination have rid Brown’s body of HIV. The first is the process of conditioning, in which doctors destroyed Brown’s own immune system with chemotherapy and whole body irradiation to prepare him for his bone marrow transplant. His oncologist, Gero Hütter, who was then with the Free University of Berlin, also took an extra step that he thought might not only cure the leukemia but also help rid Brown’s body of HIV. He found a bone marrow donor who had a rare mutation in a gene that cripples a key receptor on white blood cells the virus uses to establish an infection. (For years, researchers referred to Brown as “the Berlin patient.”) The third possibility is his new immune system attacked remnants of his old one that held HIV-infected cells, a process known as graft versus host disease.
 
In the new study, a team led by immunologist Guido Silvestri of Emory University in Atlanta, designed an unusual monkey experiment to test these possibilities.
 
Bone marrow transplants work because of stem cells. Modern techniques avoid actually aspirating bone marrow, and instead can sift through blood and pluck out the stem cells needed for a transplant to “engraft.” So the researchers first drew blood from three rhesus macaque monkeys, removed stem cells, and put the cells in storage. They then infected these animals and three control monkeys with a hybrid virus, known as SHIV, that contains parts of the simian and human AIDS viruses. All six animals soon began receiving ARVs (which respond better to SHIVs than SIV itself), and SHIV levels in the blood quickly dropped below the level of detection on standard tests, as expected.
 
A few months later, the three monkeys that had stored stem cells underwent whole body irradiation to condition their bodies and then had their own stem cells reinfused. After the cells engrafted, a process that took a few more months, the researchers stopped ARVs in the three animals and in the three controls. SHIV quickly came screaming back in the three controls and two of the transplanted animals. (One of the transplanted monkeys did not have the virus rebound but its kidneys failed and the researchers euthanized it.)
 
The team, which publishes its work online in PLOS Pathogens today, concludes that conditioning by itself likely cannot rid the body of the AIDS virus. Silvestri explains that the monkey study was a proof-of-principle experiment that cleanly isolated the effects of conditioning alone. “There’s no way to do this in humans,” he says.
 
 “It’s an important study and it’s a very useful model,” says Daniel Kuritzkes of Brigham & Women’s Hospital in Cambridge, Massachusetts, who wasn’t connected to the research.
Kuritzkes and colleagues are particularly interested in the experiment because two of their own HIV-infected patients with leukemia received bone marrow transplants from donors who did not have HIV-resistant cells. For several months after stopping ARVs, HIV remained at bay in both men, raising hopes that the resistant donor cells were not a factor. But the virus eventually returned in each patient. Kuritzkes suspects that the transplants did reduce the amount of HIV left in the patients’ bodies—known as the viral reservoir—but the virus resurfaced because it continued to copy itself and eventually overwhelmed the immune responses against it.
 
Although the study shows that conditioning by itself likely cannot eliminate an HIV infection, the study leaves open the possibility that graft versus host disease played a central role in Brown’s cure. Unlike Brown and Kuritzkes’s two patients, the transplanted monkeys received their own stem cells, which did not trigger a graft versus host response. “At the end of the day that might be an important component,” Silvestri says. He also thinks it might help reduce the reservoir size to treat monkeys with ARVs for longer than a few months.
 
Silvestri hopes to do future monkey experiments that test the different variables, including transplanting the animals with viral-resistant blood cells that mimic the ones that Brown received. “The best scientific studies raise as many questions as answers,” says Steven Deeks, a researcher and clinician at the University of California, San Francisco, who has treated and studied Brown. “Unfortunately, the heroic efforts that went into this study failed to provide a definitive answer regarding the riddles of the Berlin patient. The model will likely need to be further optimized, and at the very least, the macaques treated with antiretroviral therapy for longer periods of time. But I am confident the team will figure this out.”

By                
 
     
Jon is a staff writer for Science.
 
Related content:
Posted in Health
 
http://news.sciencemag.org/health/2014/09/how-did-berlin-patient-rid-himself-hiv

Saturday, September 27, 2014

Ebola — A Growing Threat?

The recent emergence of Zaire ebolavirus in West Africa1 has come as a surprise in a region more commonly known for its endemic Lassa fever, another viral hemorrhagic fever caused by an Old World arenavirus. Yet the region has seen previous ebolavirus activity  (see map). 
In the mid-1990s, scientists discovered Côte d'Ivoire ebolavirus (now known as Taï Forest ebolavirus) as a cause of a single reported nonfatal case in a researcher who performed a necropsy on an infected chimpanzee. The episode initiated a major research investigation in and around the Taï Forest region — an effort that failed to identify the reservoir of this new Ebola species. Since that incident, West African countries have not reported any evidence of the presence of ebolavirus.
 
Ebolaviruses belong to the family Filoviridae, a taxonomic group of enveloped, nonsegmented, negative-strand RNA viruses that includes the genera marburgvirus and cuevavirus, with a single species each, and ebolavirus, with five distinct species (see  (see figure).Bundibugyo ebolavirus to approximately 50% for Sudan ebolavirus to 70 to 90% for Zaire ebolavirus.2 The virulence of Taï Forest ebolavirus is difficult to assess because there has been only a single recorded case, and the only identified Asian species, Reston ebolavirus, seems to cause asymptomatic infection in humans.
All known African ebolaviruses can infect humans and cause similar symptoms, but they vary in terms of disease progression and virulence, with case fatality rates ranging from less than 40% for
 
Humans infected with ebolaviruses commonly present initially with nonspecific symptoms such as fever, vomiting, and severe diarrhea, with visible hemorrhage occurring in less than half the cases,2 as in the current outbreak.1 Owing to poor infrastructure, biosafety concerns associated with processes of patient care and autopsy, and the essential focus on disease containment during outbreaks, there has been little empirical study to elucidate the pathogenesis or pathology of human ebolavirus infection. The closest surrogate disease models are cynomolgus and rhesus macaques, which show clinical signs of viral hemorrhagic fever when infected with most ebolaviruses. Zaire ebolavirus is uniformly lethal in these macaques, and experts have assumed that its pathology and pathophysiology closely resemble those of ebolavirus infections in humans; immunosuppression, increased vascular permeability, and impaired coagulation have been identified as hallmarks of the disease.2 Evidence of microscopic hemorrhage is usually found, but the degree of bleeding ranges from undetectable to acutely visible. The recently introduced term “Ebola virus disease” may not convey the seriousness of a viral hemorrhagic fever, a clinical syndrome that should trigger isolation guidelines that ensure appropriate case management and implementation of infection-control measures.
 
Ebolaviruses are zoonotic pathogens purportedly carried by various species of fruit bats that are present throughout central and sub-Saharan Africa. In contrast to marburgvirus, whose reservoir has been identified as Rousettus aegyptiacus fruit bats,3 ebolaviruses have not yet been isolated from bats that have molecular and seroepidemiologic evidence of infection. Introduction into humans most likely occurs through direct contact with bats or their excretions or secretions or through contact with other end hosts, such as the great apes. Since Reston ebolavirus has been discovered in pigs on the Philippine islands, the possibility that there may be interim or amplifying hosts should not be dismissed, as we further elucidate ebolavirus ecology.
 
Human-to-human transmission leads to outbreaks, which are often started by a single introduction from the wildlife reservoir or another end host and involve virus variants with little genetic diversity, as in the current outbreak in West Africa.1 Some recorded outbreaks, on the other hand, have stemmed from multiple introductions, which have resulted in greater genetic viral diversity among the subsequent distinct chains of human-to-human transmission. Within a given species, however, virus variants have been shown to have low genetic diversity, often less than a few percent, as illustrated by the new variant isolated from patients in Guinea.1 Such limited diversity generally leads to neutralizing cross-reactivity within the species.
 
Biologic characterization of various Zaire ebolaviruses, their case fatality rates, and their virulence in animal models have so far failed to provide convincing evidence of obvious differences in pathogenicity. Thus, it should be assumed that the new West African variant is not more virulent than previous Zaire ebolaviruses; a case fatality rate of about 70%, if confirmed, might even indicate lower virulence. The finding that the Guinea variant resides at a more basal position within the clade than previously known Zaire ebolaviruses 1 argues against an introduction from Central Africa and instead supports the likelihood of distinct evolution in West Africa. These findings reinforce the hypothesis that ebolaviruses have a broader geographic distribution than previously thought.
 
There is currently no licensed prophylaxis or treatment for any ebolavirus or marburgvirus infection; therefore, treatment is merely supportive.2 Over the past decade, however, multiple countermeasure options have shown promising efficacy in macaque models of filoviruses, and some of the approaches have completed or are at least nearing phase 1 clinical trials in humans.4
 
The current front-runner for therapeutic intervention seems to be antibody treatment, which has been successful in macaques even when antibodies are administered more than 72 hours after infection. Treatment approaches involving modulatory RNA (i.e., small interfering RNAs or phosphorodiamidate morpholino oligomers) are following close behind, along with a promising synthetic drug-like small molecule, BCX4430.5 The most promising vaccine approaches are based on recombinant technologies, such as virus-like particles produced through plasmid transfection and replication-incompetent and -competent viral vectors.4 Among the latter, vesicular stomatitis virus vectors have shown efficacy within 24 to 48 hours after infection in rhesus macaques.
 
In the absence of effective intervention strategies, diagnosis becomes a key element in our response to ebolavirus infection.2 Detection rests largely on molecular techniques utilizing multiple reverse-transcriptase–polymerase-chain-reaction assays that can be used at remote outbreak sites. Antigen detection may be performed in parallel or serve as a confirmatory test for immediate diagnosis, whereas assays for detection of antibodies (e.g., IgM and IgG) are secondary tests that are primarily important in surveillance. Molecular detection strongly depends on sequence conservation, and established assays may fail when applied to new variants, strains, or viruses. Therefore, real-time sharing of information, particularly sequence data, is absolutely critical for our response capacity, since any delay could have disastrous consequences for public health. In addition, diagnostics remain essential for the time-consuming process of tracing contacts during an outbreak and for overcoming the obstacles to reintroducing survivors into their community.
 
The latest outbreak of Zaire ebolavirus in West Africa again has shown the limited ability of our public health systems to respond to rare, highly virulent communicable diseases. The medical and public health sectors urgently need to improve education and vigilance. And rapid, reliable diagnostic procedures must be implemented in key regions within or closer to the areas where these viruses are endemic so that local public health systems do not have to rely on distant reference laboratories, which should play a more confirmatory role in the future. Moreover, to optimize diagnostic-response capabilities, it is essential that information be shared in real time, as it was during the pandemic of the severe acute respiratory syndrome and during recurrent outbreaks of influenza.
 
Despite years of research on ebolaviruses and marburgviruses, it is still not possible to administer vaccines or treatments to the at-risk population or medical aid teams. If we are to practice cutting-edge medicine, rather than simply outbreak control, we need to advance leading approaches toward approval and licensing. This gap should close over the next several years — if we can continue making progress before Ebola (or a related virus) strikes again.
Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.
This article was published on May 7, 2014, and updated on May 22, 2014, at NEJM.org.

Source Information

From the Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, Hamilton, MT.


http://www.nejm.org/action/showMediaPlayer?doi=10.1056%2FNEJMp1410741&aid=NEJMp1410741_attach_1&area=
 

Friday, September 26, 2014

mRNA-based therapeutics — developing a new class of drugs

Nature Reviews Drug Discovery | Review

The therapeutic potential of in vitro-transcribed mRNA (IVT mRNA) extends from prophylactic and therapeutic vaccines to applications such as protein replacement and genome engineering. In this Review, the authors describe the recent developments in the IVT mRNA field, discuss the class-specific challenges with regards to translating IVT mRNA into a biopharmaceutical, and provide an overview of IVT mRNA drugs in development for different indications.

Key points
  • Messenger RNA (mRNA) is a pivotal molecule of life, involved in almost all aspects of cell biology.
  • As the subject of basic and applied research for more than 5 decades, mRNA has only recently come into the focus as a potentially powerful drug class able to deliver genetic information. 
  • Synthetic mRNA can be engineered to resemble mature and processed mRNA molecules as they occur naturally in the cytoplasm of eukaryotic cells and to transiently deliver proteins. 
  • Recent advances addressed challenges inherent to this drug class and provided the basis for a broad spectrum of applications 
  • Besides cancer immunotherapies and infectious disease vaccines novel approaches such as in vivo delivery of mRNA to replace or supplement proteins, mRNA-based induction of pluripotent stem cells, or mRNA-assisted delivery of designer nucleases for genome engineering rapidly emerged and entered into pharmaceutical development. 
  • This Review gives a comprehensive overview of the current state of mRNA drug technologies, their applications and crucial aspects relevant to mRNA based drug discovery and development.

Abstract:


In vitro transcribed (IVT) mRNA has recently come into focus as a potential new drug class to deliver genetic information. Such synthetic mRNA can be engineered to transiently express proteins by structurally resembling natural mRNA. Advances in addressing the inherent challenges of this drug class, particularly related to controlling the translational efficacy and immunogenicity of the IVTmRNA, provide the basis for a broad range of potential applications. mRNA-based cancer immunotherapies and infectious disease vaccines have entered clinical development. Meanwhile, emerging novel approaches include in vivo delivery of IVT mRNA to replace or supplement proteins, IVT mRNA-based generation of pluripotent stem cells and genome engineering using IVT mRNA-encoded designer nucleases. This Review provides a comprehensive overview of the current state of mRNA-based drug technologies and their applications, and discusses the key challenges and opportunities in developing these into a new class of drugs.


Figure 1: (Timeline): Key discoveries and advances in the development of mRNA as a drug technology
CAR, chimeric antigen receptor; Cas9, CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeat; DC, dendritic cell; dsRNA, double-stranded RNA; iPSC, induced pluripotent stem cell; RSV, respiratory syncytial virus; ssRNA, single stranded RNA; TALEN, transcription activator-like effector nuclease; TLR, Toll-like receptor. 

Figure 2: Principles of antigen-encoding mRNA pharmacology.
a | A linearized DNA plasmid template with the antigen-coding sequence is used for in vitro transcription. The in vitro transcribed mRNA contains the cap, 5′and 3′ untranslated regions (UTRs), the open reading frame (ORF) and the poly(A) tail, which determine the translational activity and stability of the mRNA molecule after its transfer into cells. b | Step 1: a fraction of exogenous mRNA escapes degradation by ubiquitous RNases and is spontaneously endocytosed by cell-specific mechanisms (for example, macropinocytosis in immature dendritic cells) and enters endosomal pathways. Step 2: release mechanisms of mRNA into the cytoplasm are not fully understood. Step 3: translation of mRNA uses the protein synthesis machinery of host cells. The rate-limiting step of mRNA translation is the binding of the eukaryotic translation initiation factor 4E (eIF4E) to the cap structure 222, 223. Binding of the mRNA to ribosomes, the eukaryotic initiation factors eIF4E and eIF4G, and poly(A)-binding protein, results in the formation of circular structures and active translation 224. Step 4: termination of translation by degradation of mRNAs is catalysed by exonucleases 225, 226. The cap is hydrolysed by the scavenger decapping enzymes DCP1, DCP2 and DCPS 32, followed by digestion of the residual mRNA by 5′–3′ exoribonuclease 1 (XRN1). Degradation may be delayed if the mRNA is silenced and resides in cytoplasmic processing bodies 227. Alternatively, endonucleolytic cleavage of mRNA in the exosome may occur 228, 229, 230. The catabolism of abberant mRNA (for example, mRNA with a premature stop codon) is controlled by various other mechanisms 231. Step 5: the translated protein product undergoes post-translational modification, the nature of which depends on the properties of the host cell. The translated protein can then act in the cell in which it has been generated. Step 6: alternatively, the protein product is secreted and may act via autocrine, paracrine or endocrine mechanisms. Step 7: for immunotherapeutic use of mRNA, the protein product needs to be degraded into antigenic peptide epitopes. These peptide epitopes are loaded onto major histocompatibility complex (MHC) molecules, which ensure surface presentation of these antigens to immune effector cells. Cytoplasmic proteins are proteasomally degraded and routed to the endoplasmic reticulum where they are loaded on MHC class I molecules to be presented to CD8+ cytotoxic T lymphocytes. MHC class I molecules are expressed by almost all cells. Step 8: in antigen-presenting cells, to obtain cognate T cell help for a more potent and sustainable immune response, the protein product needs to be routed to MHC class II loading compartments. This can be accomplished by incorporating routing signal-encoding sequences into the mRNA. Moreover, exogenous antigens that are taken up by dendritic cells can also be processed and loaded onto MHC class I molecules by a mechanism that is known as cross-priming 232. Step 9: protein-derived epitopes can then be presented on the cell surface by both MHC class I and MHC class II molecules. 

Figure 3: Tuning mRNA drug dose pharmacokinetics.
a | Key structural elements of in vitro transcribed (IVT) mRNA and strategies for their modifications. b | Depending on which elements (for example, modification of caps, untranslated regions (UTRs) or poly(A) tails) are used alone or in combination, the duration and kinetic profile of expression of the protein product can be modulated and fine-tuned. eIF4E, eukaryotic translation initiation factor 4E; IRES, internal ribosome entry site; ORF, open reading frame. 

Figure 4: Inflammatory responses to synthetic mRNA.
In vitro transcribed (IVT) mRNA is recognized by various endosomal innate immune receptors (Toll-like receptor 3 (TLR3), TLR7 and TLR8) and cytoplasmic innate immune receptors (protein kinase RNA-activated (PKR), retinoic acid-inducible gene I protein (RIG-I), melanoma differentiation-associated protein 5 (MDA5) and 2′–5′-oligo adenylate synthase (OAS)). Signaling through these different pathways results in inflammation associated with type 1 interferon (IFN), tumour necrosis factor (TNF), interleukin-6 (IL-6), IL-12 and the activation of cascades of transcriptional programmes. Overall, these create a pro-inflammatory microenvironment poised for inducing specific immune responses. Moreover, downstream effects such as slow-down of translation by eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, enhanced RNA degradation by ribonuclease L (RNASEL) over expression and inhibition of replication of self-amplifying mRNA are of relevance for the pharmacokinetics and pharmacodynamics of the IVT mRNA. IRF, interferon regulatory factor; ISRE7, interferon-stimulated response element; MAVS, mitochondrial antiviral signaling protein; MDA5, melanoma differentiation-associated protein 5; MYD88, myeloid differentiation primary response protein 88; MX1, myxovirus (influenza) resistance 1; NF-κB, nuclear factor-κB; TRIF, Toll-IL-1 receptor domain-containing adapter protein inducing IFNβ.

Figure 5: Differences in siRNA, pDNA and mRNA technologies in tissues with non-fenestrated or fenestrated capillaries.
All three nucleic acid-based drug modalities are applied as nanosized drug formulations for systemic delivery and reach organs via capillary systems with either non-fenestrated (a) or fenestrated (b) capillaries. The primary pharmacological effect of small interfering RNA (siRNA), namely the deletion of a defined protein function in situ, is restricted to those very cells it has entered. siRNA cannot act in cells that are not directly accessed owing to biological barriers such as non-fenestrated capillaries. In tissues with endothelial fenestration, siRNA may reach a few tissue layers adjacent to capillaries. Plasmid DNA (pDNA) is only incorporated and active in those cells undergoing mitosis at the time of exposure. This impairs its use for tissues with non-fenestrated capillaries and restricts the number of transfectable cells in tissues with endothelial fenestration to those undergoing mitosis at the time of exposure. In contrast to pDNA, mRNA enters and acts in endothelial cells of non-fenestrated tissues, and in fenestrated tissues it reaches both mitotic and non-mitotic cells in cell layers adjacent to the capillaries 233. Non-target cells, such as vascular endothelial cells transfected with mRNA or pDNA, can express pharmacologically active proteins and, via paracrine secretion, can reach target cells that are located behind the mRNA delivery barriers 234 (obviously siRNA cannot rely on such a function). Proteins produced in transfected cells are able to diffuse after secretion into the target tissue and mediate the intended biological effects via paracrine activity on adjacent cell populations. Such paracrine activity may be of particular value in tissues that have non-fenestrated capillaries.

In vitro transcribed (IVT) mRNA is recognized by various endosomal innate immune receptors (Toll-like receptor 3 (TLR3), TLR7 and TLR8) and cytoplasmic innate immune receptors (protein kinase RNA-activated (PKR), retinoic acid-inducible gene I protein (RIG-I), melanoma differentiation-associated protein 5 (MDA5) and 2′–5′-oligoadenylate synthase (OAS)). Signalling through these different pathways results in inflammation associated with type 1 interferon (IFN), tumour necrosis factor (TNF), interleukin-6 (IL-6), IL-12 and the activation of cascades of transcriptional programmes. Overall, these create a pro-inflammatory microenvironment poised for inducing specific immune responses. Moreover, downstream effects such as slow-down of translation by eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, enhanced RNA degradation by ribonuclease L (RNASEL) overexpression and inhibition of replication of self-amplifying mRNA are of relevance for the pharmacokinetics and pharmacodynamics of the IVT mRNA. IRF, interferon regulatory factor; ISRE7, interferon-stimulated response element; MAVS, mitochondrial antiviral signalling protein; MDA5, melanoma differentiation-associated protein 5; MYD88, myeloid differentiation primary response protein 88; MX1, myxovirus (influenza) resistance 1; NF-κB, nuclear factor-κB; TRIF, Toll-IL-1 receptor domain-containing adapter protein inducing IFNβ.
Figure 6: Potential therapeutic applications of IVT mRNA.
The therapeutic applications of in vitro transcribed (IVT) mRNA are summarized in detail in Table 1. The solid arrows pointing in the right hand column denote applications that are in the clinic, whereas stippled arrows refer to preclinical applications. Cas9, CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeat; EPO, erythropoietin; FOXP3, forkhead box P3; IL-10, interleukin-10; MSC, mesenchymal stem cell; RSV, respiratory syncytial virus; SPB, surfactant protein B; TALEN, transcription activator-like effector nuclease; VEGFA, vascular endothelial growth factor A; ZNF, zinc finger nuclease.

Author: Ugur Sahin, Katalin Karikó & Özlem Türeci 
Publication: Nature Reviews Drug Discovery | Review 
Publisher: Nature Publishing Group 
Date:19 September 2014 
Copyright © 2014, Rights Managed by Nature Publishing Group
 

Thursday, September 25, 2014

Noncoding RNAs and myocardial fibrosis

Nature Reviews Cardiology

 
During stress or injury-induced cardiac remodelling, fibroblasts increase production of extracellular matrix proteins, which leads to fibrosis formation, and consequently, heart failure. In this Review, Thomas Thum describes the contribution of noncoding RNAs to this process, with a specific focus on microRNAs that might be used as future therapeutic targets or biomarkers for cardiac fibrosis.

Abstract:

Cardiac stress leads to remodelling of cardiac tissue, which often progresses to heart failure and death. Part of the remodelling process is the formation of fibrotic tissue, which is caused by exaggerated activity of cardiac fibroblasts leading to excessive extracellular matrix production within the myocardium. Noncoding RNAs (ncRNAs) are a diverse group of endogenous RNA-based molecules, which include short (~22 nucleotides) microRNAs and long ncRNAs (of >200 nucleotides). These ncRNAs can regulate important functions in many cardiovascular cells types. This Review focuses on the role of ncRNAs in cardiac fibrosis; specifically, ncRNAs as therapeutic targets, factors for direct fibroblast transdifferentation, their use as diagnostic and prognostic markers, and their potential to function as paracrine modulators of cardiac fibrosis and remodelling.
 
Figure 1: ncRNAs in fibroblast biology.
miRNAs and lncRNAs are important intracellular regulators of gene expression, but also directly or indirectly regulate proteins. pri-miRNAs are processed by Drosha into pre-miRNAs, before the endonuclease Dicer generates a mature miRNA. ncRNAs serve as therapeutic targets, but are also secreted from fibroblasts and are potential diagnostic markers and paracrine signalling mediators between cells. miRNAs and lncRNAs are likely to be involved in differentitation and transdifferentiation processes, such as direct transdifferentiation of fibroblasts towards a cardiomyocyte fate. Abbreviations: lncRNA, long noncoding RNA; miRNA, microRNA; ncRNA, noncoding RNA.

Figure 2: ncRNAs in transdifferentiation of fibroblasts towards cardiomyocytes.
A combination of different factors, such as GATA-4, TBX5, and MEF2C, can initiate transdifferentiation, although these events are rare. Addition of miRNAs and possibly lncRNAs are likely to support this process. In the future, these factors might form therapeutic approaches to enable fibrotic scarring in the heart to redifferentiate into healthy functional myocardium. Whether cardiomyocytes can dedifferentiate back into fibroblasts is currently unknown. Abbreviations: GATA-4, transcription factor GATA-4; HAND2, heart and neural crest derivatives-expressed protein 2; JAK1, tyrosine-protein kinase JAK1; lncRNA, long noncoding RNA; MEF2C, myocyte-specific enhancer factor 2C; miRNA, microRNA; ncRNA, noncoding RNA; TBX5, T-box transcription factor TBX5.

Figure 3: Cardiac fibroblasts involved in intercellular communication.
Cardiac fibroblasts communicate with multiple cell types within the myocardium, such as cardiomyocytes, with endothelial and immune cells by secretion of cytokines and growth factors, and by exchanging genetic material such as miRNAs via vesicles. The communication is not unidirectional and nonfibroblast cells communicate with cardiac fibroblasts via cytokines, growth factors (such as FGF2 and CTGF), probably via vesicles, and also by direct cell–cell communication. Abbreviations: CTGF, connective tissue growth factor; FGF2, fibroblast growth factor 2; lncRNA, long noncoding RNA; miRNA, microRNA; ncRNA, noncoding RNA.
 
Title:
Author:
Thomas Thum
Publication:
Nature Reviews Cardiology
Publisher:
Nature Publishing Group
Date:
Sep 9, 2014
Copyright © 2014, Rights Managed by Nature Publishing Group

Friday, September 19, 2014

Mitochondrial dynamics in the central regulation of metabolism

Nature Reviews Endocrinology

 
Mitochondria have a fundamental role in regulating metabolic pathways and in maintaining energy balance. In this Review, Carole Nasrallah and Tamas Horvath discuss the contribution of mitochondrial function, in particular mitochondrial dynamics, to central metabolism from the hypothalamic perspective and describe how mitochondrial dysfunction can lead to the development of metabolic diseases.
 

Abstract:

The ability of an organism to convert organic molecules from the environment into energy is essential for the development of cellular structures, cell differentiation and growth. Mitochondria have a fundamental role in regulating metabolic pathways, and tight control of mitochondrial functions and dynamics is critical to maintaining adequate energy balance. In complex organisms, such as mammals, it is also essential that the metabolic demands of various tissues are coordinated to ensure that the energy needs of the whole body are effectively met. Within the arcuate nucleus of the hypothalamus, the NPY–AgRP and POMC neurons have a crucial role in orchestrating the regulation of hunger and satiety. Emerging findings from animal studies have revealed an important function for mitochondrial dynamics within these two neuronal populations, which facilitates the correct adaptive responses of the whole body to changes in the metabolic milieu. The main proteins implicated in these studies are the mitofusins, Mfn1 and Mfn2, which are regulators of mitochondrial dynamics. In this Review, we provide an overview of the mechanisms by which mitochondria are involved in the central regulation of energy balance and discuss the implications of mitochondrial dysfunction for metabolic disorders.
 
Figure 1: Melanocortin system in the arcuate nucleus of the hypothalamus.
 
Melanocortin system in the arcuate nucleus of the hypothalamus.
 
Figure 2: Regulation of POMC and NPY–AgRP neuron activation.
 
Regulation of POMC and NPY-AgRP neuron activation.
 

Figure 3: Feeding states and mitochondrial dynamics in NPY–AgRP and POMC neurons.
 

Feeding states and mitochondrial dynamics in NPY-AgRP and POMC neurons.

Title:
Mitochondrial dynamics in the central regulation of metabolism
Author:
Carole M. Nasrallah, Tamas L. Horvath
Publication:
Nature Reviews Endocrinology
Publisher:
Nature Publishing Group
Date:
Sep 9, 2014