Academic literature on the topic 'Mitochondria, rare diseases, gene therapy'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Mitochondria, rare diseases, gene therapy.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Mitochondria, rare diseases, gene therapy"

1

Ramón, Javier, Ferran Vila-Julià, David Molina-Granada, Miguel Molina-Berenguer, Maria Jesús Melià, Elena García-Arumí, Javier Torres-Torronteras, Yolanda Cámara, and Ramon Martí. "Therapy Prospects for Mitochondrial DNA Maintenance Disorders." International Journal of Molecular Sciences 22, no. 12 (June 16, 2021): 6447. http://dx.doi.org/10.3390/ijms22126447.

Full text
Abstract:
Mitochondrial DNA depletion and multiple deletions syndromes (MDDS) constitute a group of mitochondrial diseases defined by dysfunctional mitochondrial DNA (mtDNA) replication and maintenance. As is the case for many other mitochondrial diseases, the options for the treatment of these disorders are rather limited today. Some aggressive treatments such as liver transplantation or allogeneic stem cell transplantation are among the few available options for patients with some forms of MDDS. However, in recent years, significant advances in our knowledge of the biochemical pathomechanisms accounting for dysfunctional mtDNA replication have been achieved, which has opened new prospects for the treatment of these often fatal diseases. Current strategies under investigation to treat MDDS range from small molecule substrate enhancement approaches to more complex treatments, such as lentiviral or adenoassociated vector-mediated gene therapy. Some of these experimental therapies have already reached the clinical phase with very promising results, however, they are hampered by the fact that these are all rare disorders and so the patient recruitment potential for clinical trials is very limited.
APA, Harvard, Vancouver, ISO, and other styles
2

Chojdak-Łukasiewicz, Justyna, Edyta Dziadkowiak, and Sławomir Budrewicz. "Monogenic Causes of Strokes." Genes 12, no. 12 (November 23, 2021): 1855. http://dx.doi.org/10.3390/genes12121855.

Full text
Abstract:
Strokes are the main cause of death and long-term disability worldwide. A stroke is a heterogeneous multi-factorial condition, caused by a combination of environmental and genetic factors. Monogenic disorders account for about 1% to 5% of all stroke cases. The most common single-gene diseases connected with strokes are cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) Fabry disease, mitochondrial myopathy, encephalopathy, lactacidosis, and stroke (MELAS) and a lot of single-gene diseases associated particularly with cerebral small-vessel disease, such as COL4A1 syndrome, cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), and Hereditary endotheliopathy with retinopathy, nephropathy, and stroke (HERNS). In this article the clinical phenotype for the most important single-gene disorders associated with strokes are presented. The monogenic causes of a stroke are rare, but early diagnosis is important in order to provide appropriate therapy when available.
APA, Harvard, Vancouver, ISO, and other styles
3

Mishra, Ratnakar, Benson S. Chen, Prachi Richa, and Patrick Yu-Wai-Man. "Wolfram syndrome: new pathophysiological insights and therapeutic strategies." Therapeutic Advances in Rare Disease 2 (January 2021): 263300402110395. http://dx.doi.org/10.1177/26330040211039518.

Full text
Abstract:
Wolfram Syndrome (WS) is an ultra-rare, progressive neurodegenerative disease characterized by early-onset diabetes mellitus and irreversible loss of vision, secondary to optic nerve degeneration. Visual loss in WS is an important cause of registrable blindness in children and young adults and the pathological hallmark is the preferential loss of retinal ganglion cells within the inner retina. In addition to optic atrophy, affected individuals frequently develop variable combinations of neurological, endocrinological, and psychiatric complications. The majority of patients carry recessive mutations in the WFS1 (4p16.1) gene that encodes for a multimeric transmembrane protein, wolframin, embedded within the endoplasmic reticulum (ER). An increasingly recognised subgroup of patients harbor dominant WFS1 mutations that usually cause a milder phenotype, which can be limited to optic atrophy. Wolframin is a ubiquitous protein with high levels of expression in retinal, neuronal, and muscle tissues. It is a multifunctional protein that regulates a host of cellular functions, in particular the dynamic interaction with mitochondria at mitochondria-associated membranes. Wolframin has been implicated in several crucial cellular signaling pathways, including insulin signaling, calcium homeostasis, and the regulation of apoptosis and the ER stress response. There is currently no cure for WS; management remains largely supportive. This review will cover the clinical, genetic, and pathophysiological features of WS, with a specific focus on disease models and the molecular pathways that could serve as potential therapeutic targets. The current landscape of therapeutic options will also be discussed in the context of the latest evidence, including the pipeline for repurposed drugs and gene therapy. Plain language summary Wolfram syndrome – disease mechanisms and treatment options Wolfram syndrome (WS) is an ultra-rare genetic disease that causes diabetes mellitus and progressive loss of vision from early childhood. Vision is affected in WS because of damage to a specialized type of cells in the retina, known as retinal ganglion cells (RGCs), which converge at the back of the eye to form the optic nerve. The optic nerve is the fast-conducting cable that transmits visual information from the eye to the vision processing centers within the brain. As RGCs are lost, the optic nerve degenerates and it becomes pale in appearance (optic atrophy). Although diabetes mellitus and optic atrophy are the main features of WS, some patients can develop more severe problems because the brain and other organs, such as the kidneys and the bladder, are also affected. The majority of patients with WS carry spelling mistakes (mutations) in the WFS1 gene, which is located on the short arm of chromosome 4 (4p16.1). This gene is highly expressed in the eye and in the brain, and it encodes for a protein located within a compartment of the cell known as the endoplasmic reticulum. For reasons that still remain unclear, WFS1 mutations preferentially affect RGCs, accounting for the prominent visual loss in this genetic disorder. There is currently no effective treatment to halt or slow disease progression and management remains supportive, including the provision of visual aids and occupational rehabilitation. Research into WS has been limited by its relative rarity and the inability to get access to eye and brain tissues from affected patients. However, major advances in our understanding of this disease have been made recently by making use of more accessible cells from patients, such as skin cells (fibroblasts), or animal models, such as mice and zebrafish. This review summarizes the mechanisms by which WFS1 mutations affect cells, impairing their function and eventually leading to their premature loss. The possible treatment strategies to block these pathways are also discussed, with a particular focus on drug repurposing (i.e., using drugs that are already approved for other diseases) and gene therapy (i.e., replacing or repairing the defective WFS1 gene).
APA, Harvard, Vancouver, ISO, and other styles
4

Albiges, Laurence, Ronan Flippot, Nathalie Rioux-Leclercq, and Toni K. Choueiri. "Non–Clear Cell Renal Cell Carcinomas: From Shadow to Light." Journal of Clinical Oncology 36, no. 36 (December 20, 2018): 3624–31. http://dx.doi.org/10.1200/jco.2018.79.2531.

Full text
Abstract:
Non–clear cell renal cell carcinomas (RCCs) account for up to 25% of kidney cancers and encompass distinct diseases with distinct pathologic features, different molecular alterations, and various patterns of response to systemic therapies. Recent advances in molecular biology and large collaborative efforts helped to better define the oncogenic mechanisms at play in papillary, chromophobe, collecting duct, medullary, translocation, and sarcomatoid RCCs. Papillary RCCs are divided into several subsets of tumors characterized by distinct gene expression profiles, chromatin remodeling genes, cell cycle changes, and alterations of the MET pathway. Chromophobe RCC genomic analysis revealed mostly metabolic pathway alterations with mitochondrial dysfunctions. Translocation RCCs are characterized by MITF fusions and wide genomic reprogramming. Collecting duct carcinomas are distinct entities from upper tract urothelial carcinomas associated with high T-cell infiltration and metabolic alterations. Medullary RCCs present alterations of the INI1 gene and rhabdoid features at pathologic analysis. Finally, sarcomatoid RCCs represent sarcomatoid differentiation for any subsets of RCCs with specific alterations associated with mesenchymal dedifferentiation. From the standpoint of systemic therapy, more than a decade of using VEGF and mTOR inhibitors showed that they generally had limited efficacy in non–clear cell RCCs compared with clear cell RCCs. MET inhibitors are actively being developed for papillary RCC with a specific focus on MET-driven tumors. Other strategies under investigation include CDK4/6 inhibitors in tumors with cell cycle alterations and EZH2 inhibitors in RCCs with INI1 loss. The emergence of immune checkpoint inhibitors and combination strategies enlarges the spectrum of investigational treatments. Better understanding of driver and passenger alterations and better patient stratification along with dedicated clinical networks will be key to improving the management of these rare tumors.
APA, Harvard, Vancouver, ISO, and other styles
5

SAHEL, JA. "Gene therapy in rare diseases." Acta Ophthalmologica 90 (August 6, 2012): 0. http://dx.doi.org/10.1111/j.1755-3768.2012.3222.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Phillips, M. Ian, and Andrew B. Burns. "The emergence of gene therapy for rare diseases." Expert Opinion on Orphan Drugs 2, no. 11 (October 28, 2014): 1197–209. http://dx.doi.org/10.1517/21678707.2014.978284.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Wang, Wenqing, Avni Awani, Lauren Reich, Yusuke Nakauchi, Daniel Thomas, Daniel P. Dever, Matthew Porteus, and Katja G. Weinacht. "An Engineered Cell-Traceable Model of Reticular Dysgenesis in Human Hematopoietic Stem Cells Linking Metabolism and Differentiation." Blood 132, Supplement 1 (November 29, 2018): 2558. http://dx.doi.org/10.1182/blood-2018-99-117926.

Full text
Abstract:
Abstract Hematopoietic stem cell (HSC) differentiation is accompanied by a metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS) to meet the increasing energy demand during proliferation and differentiation. However, the role of mitochondrial metabolism in HSC differentiation goes beyond ATP production. Metabolites generated during mitochondrial metabolism may impact in HSC fate decisions through stable epigenetic modifications. Despite some progress in understanding mitochondrial communication during HSC development, their role in human hematopoiesis remains largely elusive, where the lack of appropriate model systems poses a major obstacle. Reticular Dysgenesis (RD), a rare and particularly severe form of severe combined immunodeficiency (SCID), offers an attractive model for studying the role of mitochondrial metabolism in hematopoiesis. RD is an autosomal recessive disease caused by biallelic mutations of the mitochondrial enzyme Adenylate Kinase 2 (AK2). AK2 catalyzes the reversible phosphorylation of adenosine monophosphate (AMP) to adenosine diphosphate (ADP), which serves as the substrate for the ATP synthase. In addition to defective lymphocyte development typical of classic SCID, RD patients also suffer from impaired myeloid development, suggestive of a global defect in hematopoiesis. In a human induced pluripotent stem cell (iPSC) model for RD, hematopoietic stem and progenitor cells (HSPCs) recapitulate a profound maturation arrest of the myeloid lineage, increased oxidative stress and an energy-depleted metabolite and transcriptional profile. We hypothesize that AK2 defects drive hematopoietic cell fate decisions through changes in metabolites that regulate the activities of DNA/histone modifying enzymes and result in stable epigenetic modifications. Methods: Since iPSCs are not suitable to model the epigenetic characteristics of definitive hematopoiesis, we developed a novel model system in which we deleted AK2 in primary human HSCs using CRISPR/Cas9 gene editing technique. We found a highly effective single-guide RNA (sgRNA) targeting the catalytic LID domain of the AK2 gene to introduce directed DNA double stranded breaks (DSBs), and use a homologous recombination (HR)-mediated dual reporter system to track and isolate cells with biallelic AK2 disruption. Results: Our single-color GFP reporter system consistently produces a >60% GFP+ population of AK2-targeted CD34+ umbilical cord blood (UCB) cells. With dual GFP/BFP reporters, we were able to achieve 6% GFP/BFP double positive cells with confirmed biallelic AK2 knock-out. Since HR events on one allele are biologically linked to CRISPR/Cas9 mediated DSBs on the other, we assessed insertion and/or deletion (INDEL) frequency and protein expression in a single reporter (GFP+) population of AK2-targeted UCBs. We detected an INDEL frequency of over 90% on the non-HR alleles along with nearly absent AK2 protein expression by Western Blot. These results indicated that the highly efficient single-color reporter system with >60% targeting efficiency is sufficient to achieve an AK2 biallelic knock-out population in primary HSCs. in vitro myeloid differentiation of these AK2-targeted HSCs recapitulates the RD phenotype with impaired neutrophil but preserved monocyte development. Conclusion: This novel disease model for RD will now allow us to examine the cellular and molecular impact of perturbations in metabolism on human HSC development. We will investigate the effect on differentiation potential, metabolite profile, transcriptome and epigenome in vitro as well as in a xenograft mouse model. Elucidating how metabolism governs differentiation and self-renewal of human HSCs will not only advance our basic understanding of many blood and immune diseases, but has important translational implications for improving the use of HSCs in hematopoietic stem cell transplantation, gene and cell therapy. Disclosures Porteus: CRISPR Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees.
APA, Harvard, Vancouver, ISO, and other styles
8

Girach, Aniz. "The Future of Gene Therapy for Rare Eye Diseases." Cell and Gene Therapy Insights 4, no. 7 (October 15, 2018): 725–31. http://dx.doi.org/10.18609/cgti.2018.073.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Faria, Rúben, Prisca Boisguérin, Ângela Sousa, and Diana Costa. "Delivery Systems for Mitochondrial Gene Therapy: A Review." Pharmaceutics 15, no. 2 (February 8, 2023): 572. http://dx.doi.org/10.3390/pharmaceutics15020572.

Full text
Abstract:
Mitochondria are membrane-bound cellular organelles of high relevance responsible for the chemical energy production used in most of the biochemical reactions of cells. Mitochondria have their own genome, the mitochondrial DNA (mtDNA). Inherited solely from the mother, this genome is quite susceptible to mutations, mainly due to the absence of an effective repair system. Mutations in mtDNA are associated with endocrine, metabolic, neurodegenerative diseases, and even cancer. Currently, therapeutic approaches are based on the administration of a set of drugs to alleviate the symptoms of patients suffering from mitochondrial pathologies. Mitochondrial gene therapy emerges as a promising strategy as it deeply focuses on the cause of mitochondrial disorder. The development of suitable mtDNA-based delivery systems to target and transfect mammalian mitochondria represents an exciting field of research, leading to progress in the challenging task of restoring mitochondria’s normal function. This review gathers relevant knowledge on the composition, targeting performance, or release profile of such nanosystems, offering researchers valuable conceptual approaches to follow in their quest for the most suitable vectors to turn mitochondrial gene therapy clinically feasible. Future studies should consider the optimization of mitochondrial genes’ encapsulation, targeting ability, and transfection to mitochondria. Expectedly, this effort will bring bright results, contributing to important hallmarks in mitochondrial gene therapy.
APA, Harvard, Vancouver, ISO, and other styles
10

Seibel, Peter, Jörg Trappe, Gaetano Villani, Thomas Klopstock, Sergio Papa, and Heinz Reichmann. "Transfection of mitochondria: strategy towards a gene therapy of mitochondrial DNA diseases." Nucleic Acids Research 23, no. 1 (1995): 10–17. http://dx.doi.org/10.1093/nar/23.1.10.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Mitochondria, rare diseases, gene therapy"

1

BOTTANI, EMANUELA. "Mitochondrial diseases: from gene function to therapy." Doctoral thesis, Università degli Studi di Milano-Bicocca, 2015. http://hdl.handle.net/10281/94380.

Full text
Abstract:
Le malattie mitocondriali sono disturbi genetici caratterizzati da difetti di fosforilazione ossidativa causati da mutazioni nel DNA mitocondriale, o in geni nucleari i cui prodotti sono legati alla fosforilazione ossidativa o alla biologia mitocondriale. La prima parte del progetto è stata focalizzata sulla generazione e la caratterizzazione di un modello murino di malattia mitocondriale, Ttc19ko. I pazienti con mutazioni in TTC19 sviluppano danni neurologici e deficit di complesso III. Ttc19 è una proteina mitocondriale che sembra essere coinvolta nell’assemblaggio e/o stabilità del complesso III. Abbiamo dimostrato che il modello murino Ttc19ko ha sintomi neurologici, debolezza muscolare e riduzione dell’attività locomotoria spontanea, in analogia con la malattia umana. L’analisi istologica ha rivelato alcune anomalie neurologiche con presenza di accumuli di ubiquitina e GFAP. I topi Ttc19ko hanno una riduzione complessiva del metabolismo energetico, una diminuzione del consumo di O2 e di produzione di CO2. L’attività enzimatica del complesso III è significativamente ridotta nei tessuti e ciò è legato ad un aumento della produzione di ROS. L’analisi BNGE ha mostrato una riduzione della incorporazione della subunità catalitica Rieske nel complesso assemblato. L’immunoprecipitazione di TTC19-Flag in colture cellulari trattate con amminoacidi marcati ha rivelato una maggiore interazione tra Ttc19 e le subunità del pre-complesso III, e una minore interazione con proteine Rieske e Uqcrh, entrambe assemblate tardivamente. Abbiamo inoltre dimostrato che Ttc19 rimane associata al complesso III assemblato. Nell’insieme, questi risultati suggeriscono che Ttc19 è un fattore intrinseco di assemblaggio del complesso III che interagisce con il pre-complesso III facilitando così l'incorporazione della proteina Rieske. La seconda parte del progetto è stata focalizzata sulla messa a punto di una terapia genica su un altro modello murino di malattia mitocondriale, MPV17ko. Mutazioni in MPV17 causano una sindrome epatocerebrale con deplezione del mtDNA, insufficienza epatica a esordio precoce, gravi crisi ipoglicemiche e morte. Il trapianto di fegato e l'alimentazione frequente a base di carboidrati a lento rilascio sono le uniche terapie disponibili, anche se in seguito si sviluppano danni neurologici. Il ruolo fisiologico di MPV17 non è chiaro. Abbiamo dimostrato che MPV17 fa parte di un complesso ad alto peso molecolare a composizione sconosciuta, che è essenziale per il mantenimento del mtDNA nel fegato. In dieta standard, il topo MPV17-/- non mostra quasi alcun sintomo di disfunzione epatica, ma una dieta chetogenica porta questi animali a sviluppare cirrosi e insufficienza epatica grave. Tuttavia, quando l'espressione di MPV17 è ripristinata dalla somministrazione di virus adeno-associato, si assiste ad un ricostituzione del complesso supramolecolare contenente Mpv17, ad un ripristino completo del numero di copie di mtDNA, ed alla prevenzione dell’insufficienza epatica indotta dal dieta chetogenica. Questi risultati aprono nuove prospettive terapeutiche per il trattamento delle sindromi da deplezione del mtDNA indotte da mutazioni nel gene MPV17.
Mitochondrial diseases are genetic disorders characterized by defects in oxidative phosphorylation caused by mutations in mitochondrial DNA, or in nuclear genes whose products are related to oxidative phosphorylation or mitochondrial biology. The first part of the project was focused on the generation and characterization of a mouse model of mitochondrial disease, Ttc19ko. Patients with mutations in TTC19 were characterized by neurological impairments and mitochondrial respiratory complex III deficiency. Ttc19 is a mitochondrial protein that seems to be associated to complex III assembly and/or stability. We showed that Ttc19ko mice have neurological symptoms, muscular weakness and reduction in spontaneous locomotors activity, clearly resembling the human disease. Brain also had neurological abnormalities with presence of ubiquitin and GFAP positive staining. Comprehensive lab animals monitoring system revealed a reduction in O2 consumption, CO2 production and energy expenditure in Ttc19ko mice, indicating an overall reduction of energy metabolism. Complex III activity was significantly reduced in tissues and this was linked to an increased ROS production. BNGE analysis of mitochondrial complex III showed a substantial reduction of the incorporation of the catalytic Rieske iron-sulfur protein into the fully assembled complex. A stable isotope labelling by amino acids in cell culture (SILAC) expressing TTC19-Flag followed by immunoprecipitation and mass spec analysis revealed a higher scored interaction between Ttc19 and the subunits of the pre-complexIII, and a lower scored interaction with Rieske protein and Uqcrh, both of them are late assembled subunits. We also demonstrated that Ttc19 is associated to the fully assembled complex III. Taken together, these results suggests that Ttc19 is an intrinsic assembly factor of complex III that interacts with the pre-complex III thus facilitating the incorporation of the late assembled Rieske protein. The second part of the project was focused on a gene therapy approach on a second mouse model of mitochondrial disease, MPv17ko. Mutations in hMPV17 cause a hepatocerebral form of mtDNA depletion syndrome hallmarked by early-onset liver failure, leading to premature death. Liver transplantation and frequent feeding using slow-release carbohydrates are the only available therapies, although surviving patients develop slowly progressive neuropathy. The physiological role of Mpv17 is still unclear. We showed that Mpv17 is part of a high molecular weight complex of unknown composition, which is essential for mtDNA maintenance in liver. On a standard diet, Mpv17ko mouse shows hardly any symptom of liver dysfunction, but a ketogenic diet leads these animals to liver cirrhosis and failure. However, when expression of human MPV17 is carried out by adeno-associated virus mediated gene replacement, the Mpv17ko mice are able to reconstitute the Mpv17-containing supramolecular complex, restore liver mtDNA copy number and oxidative phosphorylation proficiency and prevent liver failure induced by the KD. These results open new therapeutic perspectives for the treatment of MPV17-related liver-specific MDS.
APA, Harvard, Vancouver, ISO, and other styles
2

Hidaka, Takuya. "Development of Sequence-Specific DNA Binders for the Therapy of Mitochondrial Diseases." Doctoral thesis, Kyoto University, 2021. http://hdl.handle.net/2433/263495.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Foster, Robert Graham. "Development of a modular in vivo reporter system for CRISPR-mediated genome editing and its therapeutic applications for rare genetic respiratory diseases." Thesis, University of Edinburgh, 2018. http://hdl.handle.net/1842/33040.

Full text
Abstract:
Rare diseases, when considered as a whole, affect up to 7% of the population, which would represent 3.5 million individuals in the United Kingdom alone. However, while 'personalised medicine' is now yielding remarkable results using recent sequencing technologies in terms of diagnosing genetic conditions, we have made much less headway in translating this patient information into therapies and effective treatments. Even with recent calls for greater research into personalised treatments for those affected by a rare disease, progress in this area is still severely lacking, in part due to the astronomical cost and time involved in bringing treatments to the clinic. Gene correction using the recently-described genome editing technology CRISPR/Cas9, which allows precise editing of DNA, offers an exciting new avenue of treatment, if not cure, for rare diseases; up to 80% of which have a genetic component. This system allows the researcher to target any locus in the genome for cleavage with a short guide-RNA, as long as it precedes a highly ubiquitous NGG sequence motif. If a repair sequence is then also provided, such as a wild-type copy of the mutated gene, it can be incorporated by homology-directed repair (HDR), leading to gene correction. As both guide-RNA and repair template are easily generated, whilst the machinery for editing and delivery remain the same, this system could usher in the era of 'personalised medicine' and offer hope to those with rare genetic diseases. However, currently it is difficult to test the efficacy of CRISPR/Cas9 for gene correction, especially in vivo. Therefore, in my PhD I have developed a novel fluorescent reporter system which provides a rapid, visual read-out of both non-homologous end joining (NHEJ) and homology-directed repair (HDR) driven by CRISPR/Cas9. This system is built upon a cassette which is stably and heterozygously integrated into a ubiquitously expressed locus in the mouse genome. This cassette contains a strong hybrid promoter driving expression of membrane-tagged tdTomato, followed by a strong stop sequence, and then membrane-tagged EGFP. Unedited, this system drives strong expression of membrane-tdTomato in all cell types in the embryo and adult mouse. However, following the addition of CRISPR/Cas9 components, and upon cleavage, the tdTomato is rapidly excised, resulting via NHEJ either in cells without fluorescence (due to imperfect deletions) or with membrane-EGFP. If a repair template containing nuclear tagged-EGFP is also supplied, the editing machinery may then use the precise HDR pathway, which results in a rapid transition from membrane-tdTomato to nuclear- EGFP. Thereby this system allows the kinetics of editing to be visualised in real time and allows simple scoring of the proportion of cells which have been edited by NHEJ or corrected by HDR. It therefore provides a simple, fast and scalable manner to optimise reagents and protocols for gene correction by CRISPR/Cas9, especially compared to sequencing approaches, and will prove broadly useful to many researchers in the field. Further to this, I have shown that methods which lead to gene correction in our reporter system are also able to partially repair mutations found in the disease-causing gene, Zmynd10; which is implicated in the respiratory disorder primary ciliary dyskinesia (PCD), for which there is no effective treatment. PCD is an autosomal-recessive rare disorder affecting motile cilia (MIM:244400), which results in impaired mucociliary clearance leading to neonatal respiratory distress and recurrent airway infections, often progressing to lung failure. Clinically, PCD is a chronic airway disease, similar to CF, with progressive deterioration of lung function and lower airway bacterial colonization. However, unlike CF which is monogenic, over 40 genes are known to cause PCD. The high genetic heterogeneity of this rare disease makes it well suited to such a genome editing strategy, which can be tailored for the correction of any mutated locus.
APA, Harvard, Vancouver, ISO, and other styles
4

Lu, Keyin. "Ischemic preconditioning and hydrodynamic delivery for the prevention of acute kidney injury." Thesis, 2015. http://hdl.handle.net/1805/7966.

Full text
Abstract:
Indiana University-Purdue University Indianapolis (IUPUI)
Acute Kidney Injury (AKI) is a prevalent and significant problem whose primary treatment is supportive care. Ischemic preconditioning is a strategy used to protect organs from ischemic injury via a prior injury. Ischemic preconditioning in the kidneys has been shown to confer protection onto kidneys from subsequent ischemic insults with attenuated serum creatinine values in treated rats. In the preconditioned kidneys, the enzyme IDH2 was discovered to be upregulated in the mitochondria. Hydrodynamic fluid delivery to the kidney was found to be a viable technique for delivering this gene to the kidney, resulting in artificially upregulated expression of IDH2. Via a two-pronged effort to discern the functional significance of ischemic preconditioning and hydrodynamic IDH2 fluid injections, we performed mitochondrial oxygen respiration assays on both preconditioned and injected kidneys. We found that renal ischemic preconditioning resulted in no significant difference between sham and preconditioned, subsequently injured kidneys, which is similar to the results from the serum creatinine studies. Hydrodynamically IDH2-injected, and subsequently injured kidneys respire significantly better than vehicle injected, and subsequently injured kidneys, which shows that hydrodynamic injections of IDH2 protects kidneys against injury, and partially mimics the effects of preconditioning.
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Mitochondria, rare diseases, gene therapy"

1

G, Thoene Jess, ed. Small molecule therapy for genetic disease. Cambridge: Cambridge University Press, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
2

Thoene, Jess G. Small Molecule Therapy for Genetic Disease. Cambridge University Press, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
3

Thoene, Jess G. Small Molecule Therapy for Genetic Disease. Cambridge University Press, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Thoene, Jess G. Small Molecule Therapy for Genetic Disease. Cambridge University Press, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
5

Thoene, Jess G. Small Molecule Therapy for Genetic Disease. Cambridge University Press, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
6

Schwartz, Peter J., and Lia Crotti. Monogenic and oligogenic cardiovascular diseases: genetics of arrhythmias—catecholaminergic polymorphic ventricular tachycardia. Oxford University Press, 2018. http://dx.doi.org/10.1093/med/9780198784906.003.0152.

Full text
Abstract:
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a rare inherited disorder associated with syncope and sudden death manifesting in the young during sympathetic activation. The electrocardiogram is normal and the heart is structurally normal. The diagnosis is usually made with an exercise stress test that shows a typical pattern of onset and offset of adrenergically induced ventricular arrhythmias. Molecular screening of RyR2, the major CPVT gene, is recommended whenever the suspicion of CPVT is high. If a disease-causing mutation is identified, cascade screening allows pre-symptomatic diagnosis among family members. All affected subjects should be treated with beta blockers (nadolol or propranolol). Preliminary data support the association of beta blockers with flecainide. After a cardiac arrest, an implantable cardioverter defibrillator (ICD) should be implanted, but it is accompanied by a disquietingly high incidence of adverse effects. After syncope on beta blocker therapy, left cardiac sympathetic denervation is most effective, preserves quality of life, and does not preclude a subsequent ICD implantation.
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Mitochondria, rare diseases, gene therapy"

1

Chow, Shein-Chung. "Gene Therapy for Rare Diseases." In Innovative Methods for Rare Disease Drug Development, 247–64. Boca Raton, FL : CRC Press, 2021. |: Chapman and Hall/CRC, 2020. http://dx.doi.org/10.1201/9781003049364-13.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Robinson, Peter. "Cell and Gene Therapy in Rare Diseases." In Rare Disease Drug Development, 249–61. Cham: Springer International Publishing, 2021. http://dx.doi.org/10.1007/978-3-030-78605-2_16.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Grosios, Konstantina, Harald Petry, and Jacek Lubelski. "Adeno-Associated Virus Gene Therapy and Its Application to the Prevention and Personalised Treatment of Rare Diseases." In Rare Diseases, 131–57. Dordrecht: Springer Netherlands, 2014. http://dx.doi.org/10.1007/978-94-017-9214-1_9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

López-Márquez, Arístides, Ainhoa Martínez-Pizarro, Belén Pérez, Eva Richard, and Lourdes R. Desviat. "Modeling Splicing Variants Amenable to Antisense Therapy by Use of CRISPR-Cas9-Based Gene Editing in HepG2 Cells." In Methods in Molecular Biology, 167–84. New York, NY: Springer US, 2022. http://dx.doi.org/10.1007/978-1-0716-2010-6_10.

Full text
Abstract:
AbstractThe field of splice modulating RNA therapy has gained new momentum with FDA approved antisense-based drugs for several rare diseases. In vitro splicing assays with minigenes or patient-derived cells are commonly employed for initial preclinical testing of antisense oligonucleotides aiming to modulate splicing. However, minigenes do not include the full genomic context of the exons under study and patients’ samples are not always available, especially if the gene is expressed solely in certain tissues (e.g. liver or brain). This is the case for specific inherited metabolic diseases such as phenylketonuria (PKU) caused by mutations in the liver-expressed PAH gene.Herein we describe the generation of mutation-specific hepatic cellular models of PKU using CRISPR/Cas9 system, which is a versatile and easy-to-use gene editing tool. We describe in detail the selection of the appropriate cell line, guidelines for design of RNA guides and donor templates, transfection procedures and growth and selection of single-cell colonies with the desired variant, which should result in the accurate recapitulation of the splicing defect.
APA, Harvard, Vancouver, ISO, and other styles
5

Piñón Hofbauer, Josefina, Verena Wally, Christina Guttmann-Gruber, Iris Gratz, and Ulrich Koller. "Therapy Development for Epidermolysis Bullosa." In Rare Diseases [Working Title]. IntechOpen, 2021. http://dx.doi.org/10.5772/intechopen.97437.

Full text
Abstract:
Although rare genodermatoses such as Epidermolysis bullosa have received more attention over the last years, no approved treatment options targeting causal mutations are currently available. Still, such diseases can be devastating, in some cases even associated with life-threatening secondary manifestations. Therefore, developing treatments that target disease-associated complications along with causal therapies remains the focus of current research efforts, in order to increase patient’s quality of life and potentially their life expectancy. Epidermolysis bullosa is a genodermatosis that is caused by mutations in either one of 16 genes, predominantly encoding structural components of the skin and mucosal epithelia that are crucial to give these barrier organs physical and mechanical resilience to stress. The genetic heterogeneity of the disease is recapitulated in the high variability of phenotypic expressivity observed, ranging from minor and localized blistering to generalized erosions and wound chronification, rendering certain subtypes a systemic disease that is complicated by a plethora of secondary manifestations. During the last decades, several studies have focused on developing treatments for EB patients and significant progress has been made, as reflected by numerous publications, patents, and registered trials available. Overall, strategies range from causal to symptom-relieving approaches, and include gene, RNA and cell therapies, as well as drug developments based on biologics and small molecules. In this chapter, we highlight the most recent and promising approaches that are currently being investigated in order to provide effective treatments for patients with epidermolysis bullosa in the future.
APA, Harvard, Vancouver, ISO, and other styles
6

Nita, Magdalena, Jacek Pliszczyński, Andrzej Eljaszewicz, Marcin Moniuszko, Tomasz Ołdak, Katarzyna Woźniak, Sławomir Majewski, et al. "Surgical Treatment of Wounds Using Stem Cells in Epidermolysis Bullosa (EB)." In Rare Diseases [Working Title]. IntechOpen, 2021. http://dx.doi.org/10.5772/intechopen.97036.

Full text
Abstract:
Epidermolysis bullosa (EB) is a group of hereditary skin diseases, or genodermatoses, characterized by the formation of severe, chronic blisters with painful and life-threatening complications. Despite the previous and ongoing progress in the field, there are still no effective causative treatments for EB. The treatment is limited to relieving symptoms, which—depending on disease severity—may involve skin (blisters, poorly healing wounds caused by the slightest mechanical stimuli, contractures, scarring, pseudosyndactyly) and internal organ abnormalities (esophageal, pyloric, or duodenal atresia; renal failure; and hematopoietic abnormalities). The last decade saw a series of important discoveries that paved the way for new treatment methods, including gene therapy, bone marrow transplantation, cell therapy (allogenic fibroblasts, mesenchymal stem cells [MSCs], and clinical use of induced pluripotent stem cells. Tissue engineering experts are attempting to develop skin-like structures that can facilitate the process of healing to promote skin reconstruction in injuries that are currently incurable. However, this is incredibly challenging, due to the complex structure and the many functions of the skin. Below, we characterize EB and present its potential treatment methods. Despite the cure for EB being still out of reach, recent data from animal models and initial clinical trials in humans have raised patients’, clinicians’, and researchers’ expectations. Consequently, modifying the course of the disease and improving the quality of life have become possible. Moreover, the conclusions drawn based on EB treatment may considerably improve the treatment of other genetic diseases.
APA, Harvard, Vancouver, ISO, and other styles
7

Gorodetskiy, Vadim. "Felty’s Syndrome." In Rare Diseases [Working Title]. IntechOpen, 2021. http://dx.doi.org/10.5772/intechopen.97080.

Full text
Abstract:
Felty’s syndrome (FS) is an uncommon subset of seropositive rheumatoid arthritis (RA) complicated by neutropenia with or without splenomegaly. The pathogenesis of neutropenia in FS is still not fully understood, but it is believed that the principal cause is neutrophil survival defect. Autoantibodies against peptidylarginine deiminase type 4 deiminated histones, glucose-6-phosphate isomerase, and eukaryotic elongation factor 1A-1 antigen may contribute to neutropenia development in FS patients. Splenic histology in FS shows non-specific findings and spleen size do not correlate with neutropenia. Cases of T-cell large granular lymphocytic leukemia with low tumor burden in blood and concomitant RA are clinically indistinguishable from FS and present a diagnostic challenge. Examination of T-cell clonality, mutations in signal transducer and activator of transcription 3 gene, and the number of large granular lymphocytes in the blood can establish a correct diagnosis. Optimal approaches to therapy for FS have not been developed, but the use of rituximab seems promising. In this chapter, the epidemiology, pathogenesis, clinical manifestations, differential diagnosis, and treatment options for FS are discussed.
APA, Harvard, Vancouver, ISO, and other styles
8

Tariq, Muhammad, Naveed Altaf Malik, Ilyas Ahmad, Syeda Seema Waseem, and Shahid Mahmood Baig. "Genetic Testing for Rare Genetic Disorders." In Omics Technologies for Clinical Diagnosis and Gene Therapy: Medical Applications in Human Genetics, 14–28. BENTHAM SCIENCE PUBLISHERS, 2022. http://dx.doi.org/10.2174/9789815079517122010005.

Full text
Abstract:
Rare genetic disorders affect a significant proportion of the global population. A large number of these patients are either misdiagnosed or remain undiagnosed which can have potentially adverse effects, including failure to provide anticipatory prognosis and identify potential treatment. With the completion of HGP, genetic testing has fast grown into a diagnostic discipline introducing new and cost.effective diagnostic tests with reasonable accuracy and specificity. NGS technologies, in particular, changed the field of genetic diagnosis by sequencing the entire genome or subset thereof in a single test and accomplishing diagnosis of virtually all diseases, either congenital or late-onset. These technologies have opened up new opportunities and unique challenges. This chapter discusses the importance of genetic testing, its scope, various technologies and approaches and, finally, the opportunities and challenges accompanying the new age genetic tests.
APA, Harvard, Vancouver, ISO, and other styles
9

Kazmi, Hira, and Muhammad Ilyas. "Next-Generation Technologies for Rare Inherited Disorders." In Omics Technologies for Clinical Diagnosis and Gene Therapy: Medical Applications in Human Genetics, 1–13. BENTHAM SCIENCE PUBLISHERS, 2022. http://dx.doi.org/10.2174/9789815079517122010004.

Full text
Abstract:
Rare inherited disorders have become a major public health concern in recent years. Owing to a lack of resources, poorly planned primary and basic health care, and inadequate political structures, treatment, and management policies are daunting challenges in many countries. As a result, these diseases need particular attention, especially in less developed areas, where these disorders remain unnoticed. Similarly, the effect of such severe disorders on underprivileged populations is expected to be devastating. Identifying certain genetic markers can provide a valuable explanation for disease etiology, molecular characterization, and pathogenesis. In this chapter, we highlight the importance of next-generation sequencing to explore and recognize the role of novel causative genes in developing successful treatments for the most prevalent rare genetic disorders. DNA methylation and transcriptome markers have been shown to aid in the prediction of common diseases; however, this has not been tested on rare genetic disorders. Since the rate of rare inherited disorders is higher in developing countries, we believe that these populations can provide us with much stronger clues for the genetic and environmental association. These markers, along with other parameters, can be used to systematically build machine learning models to improve risk prediction; this approach has the potential to reshape how we predict disease risk and save many lives around the world.
APA, Harvard, Vancouver, ISO, and other styles
10

Hassan, Muhammad Jawad, Muhammad Faheem, and Sabba Mehmood. "Emerging OMICS and Genetic Disease." In Omics Technologies for Clinical Diagnosis and Gene Therapy: Medical Applications in Human Genetics, 93–113. BENTHAM SCIENCE PUBLISHERS, 2022. http://dx.doi.org/10.2174/9789815079517122010010.

Full text
Abstract:
Multiomics also described as integrative omics is an analytical approach that combines data from multiple ‘omics’ approaches including genomics, transcriptomics, proteomics, metabolomics, epigenomics, metagenomics and Meta transcriptomics to answer the complex biological processes involved in rare genetic disorders. This omics approach is particularly helpful since it identifies biomarkers of disease progression and treatment progress by collective characterization and quantification of pools of biological molecules within and among the various types of cells to better understand and categorize the Mendelian and non- Mendelian forms of rare diseases. As compared to studies of a single omics type, multi-omics offers the opportunity to understand the flow of information that underlies the disease. A range of omics software and databases, for example WikiPathways, MixOmics, MONGKIE, GalaxyP, GalaxyM, CrossPlatform Commander, and iCluster are used for multi-omics data exploration and integration in rare disease analysis. Recent advances in the field of genetics and translational research have opened new treatment avenues for patients. The innovation in the next generation sequencing and RNA sequencing has improved the ability from diagnostics to detection of molecular alterations like gene mutations in specific disease types. In this chapter, we provide an overview of such omics technologies and focus on methods for their integration across multiple omics layers. The scrupulous understanding of rare genetic disorders and their treatment at the molecular level led to the concept of a personalized approach, which is one of the most significant advancements in modern research which enable researchers to better comprehend the flow of knowledge which underpins genetic diseases.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography