Flyer

International Journal of Drug Development and Research

  • ISSN: 0975-9344
  • Journal h-index: 49
  • Journal CiteScore: 11.20
  • Journal Impact Factor: 8.24
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • Genamics JournalSeek
  • China National Knowledge Infrastructure (CNKI)
  • CiteFactor
  • Scimago
  • Directory of Research Journal Indexing (DRJI)
  • OCLC- WorldCat
  • Publons
  • MIAR
  • University Grants Commission
  • Euro Pub
  • Google Scholar
  • J-Gate
  • SHERPA ROMEO
  • Secret Search Engine Labs
  • ResearchGate
  • International Committee of Medical Journal Editors (ICMJE)
Share This Page

Editorial - (2022) Volume 14, Issue 6

The Use of Animal Models in Translational Medicine, the Efficacy of Translational Research in Toxicology, Translational Breast Cancer Research, and Mitochondrial Translational

Wen-Hwa Lee*
 
1Department of Biological Chemistry, School of Medicine, University of California, Irvine CA 92697, USA
 
*Correspondence: Wen-Hwa Lee, Department of Biological Chemistry, School of Medicine, University of California, Irvine CA 92697, USA, Email:

Received: 01-Jun-2022, Manuscript No. IJDDR-22-12807; Editor assigned: 03-Jun-2022, Pre QC No. IJDDR-22-12807; Reviewed: 25-Jun-2022, QC No. IJDDR-22-12807; Revised: 30-Jun-2022, Manuscript No. IJDDR-22-12807; Published: 08-Jul-2022, DOI: 10.36648/1791-809X.22.16.959

Editorial

Translational Research

An important mediator of gene expression that is increasingly understood is translational regulation. It gives cells the capacity to choose when to express a specific protein, ensuring appropriate and timely cellular responses to environmental signals. In complicated changing contexts, such as during embryonic development, wound healing, and environmental stress, this capacity to reprogram protein synthesis and to enable the translation of the relevant regulatory instructions is particularly crucial. It should come as no surprise that errors in this mechanism might cause cancer. This review will concentrate on the translational control mechanisms present in both healthy and malignant cells. We examine the idea that a change from capdependent to cap-independent signalling may contribute to the development of primary epithelial tumours into a motile mesenchymal-like phenotype during the invasive phase of metastasis. The mitochondria use phosphorylation to convert ADP to ATP to produce the vast majority of the energy required for cellular function. The formation of free radicals and apoptosis are two additional crucial mitochondrial processes. Numerous chemical substances, including those included in the current pharmacopoeia, have the potential to affect mitochondrial function. [1] The inner mitochondrial membrane (IMM), which has electrochemical properties, the dual genetic control of mitochondrial DNA (mtDNA) and nuclear DNA (nDNA), as well as the inherent characteristics of the translational and transcriptional machinery, make the mitochondria vulnerable to environmental damage. Since mitochondria evolved from alpha-proteobacteria, the mtDNA genes are still structurally similar to the bacterial translational machinery, making them susceptible to inhibition by routinely used antibiotics that target translation. [2] There are cases where certain gene mutations may lead to higher drug toxicity; however many of these drugs have side effects in otherwise healthy people. Personalized genomic medicine and preclinical pharmacogenetic and functional studies of mitochondrial toxicity are expected to advance our knowledge of the range of diseases induced by mitochondrial translation inhibition as well as the safe and efficient application of antibiotics that inhibit bacterial and human mitochondrial translation.[3] A Special Issue titled Mitochondrial Gene Expression contains this article. Mammalian genomes are mostly transcribed. As a result, a wide variety of transcripts are produced, including long noncoding RNAs, messenger RNAs that code for proteins, and repetitive sequences like SINEs (short interspersed nuclear elements). The function of many ncRNAs, which are nuclearenriched, is uncertain. In order to control epigenetic silencing, transcription, and mRNA stability, antisense lncRNAs may link with a protein-coding gene on the opposing strand to form senseantisense pairs. Here, we identified a mouse ubiquitin carboxyterminal hydrolase L1 (Uchl1) nuclear-enriched lncRNA antisense, a gene involved in brain function and neurodegenerative disorders. A new functional class of lncRNAs has been discovered as a result of antisense Uchl1's rise in UCHL1 protein production at the post-transcriptional stage. An embedded inverted SINEB2 motif and a 5' overlapping sequence are required for antisense Uchl1 activity.[4] Other natural antisense transcripts also share these characteristics, and a manmade antisense to the green fluorescent protein can acquire regulatory action as a result. Inhibition of mTORC1 by rapamycin results in an increase in UCHL1 protein, which is linked to the movement of antisense Uchl1 RNA from the nucleus to the cytoplasm, demonstrating that the function of antisense Uchl1 is regulated by stress signalling pathways. After that, attachment of the overlapping sense protein-coding mRNA to active polysomes for translation necessitates the presence of antisense Uchl1 RNA.[5] Despite the human genome project and other molecular biology methods having revealed a huge number of possible novel medication targets, the success rates for pharmaceuticals during clinical development remain low. The development of new biological drug formats, such as Nanobodies, and advancements in the design of small molecule medications, such as computer-aided drug design, have also led to a wealth of promising therapeutic candidates that target these prospective new targets. As a result, more medications that interact with less well-validated targets have entered clinical development over the past ten years.[6] However, as a result, the success rates for medications throughout clinical development have decreased. Phases II and III of clinical development's lack of efficacy are the primary causes of these failures. Having stricter success criteria during the non-clinical stages of the drug development process could reduce the potential failure rates. This is particularly true for a given research candidate during target validation and preclinical proof of concept. The choice of animal models that enable evaluation of target validity and that can forecast clinical efficacy of a particular chemical is crucial to success in these stages. A model is a straightforward illustration of an intricate system. [7] As a result, an animal model for a human disease does not aim to replicate the complexity of the human disease in an animal, but rather to simulate particular disease-related characteristics. Therefore, it is crucial to specify a clear research question and make sure the model of choice is appropriate before utilizing it. Animal models, when properly planned and carried out, can greatly advance our understanding of biology and medicine, including the discovery and creation of new medications. Better planning, execution, and animal model development are nonetheless required. The current research addresses a few elements for enhancing the animal model's translational value.[8] In order to ensure that innovative treatments and research-based knowledge are properly applied and actually reach the patients or groups for which they are intended, the process of translating research into practise is referred to as translational research. A popular endpoint for "bench to bedside" translational research is the creation of a new medicine, although this is just the beginning for this field of study. By enhancing access, restructuring and coordinating care delivery systems, assisting clinicians and patients in changing behaviours and making more informed decisions, offering reminders and point-of-care decision support tools, and fortifying the patient-clinician relationship, translational research aims to close that gap and improve quality.[9] There are several compounds found in the toxins produced by animals, plants, and microorganisms that could be used to cure human and animal ailments. Oncology because it affects distant metastases, local control of the disease is necessary for a satisfactory treatment outcome. According to this viewpoint, breast cancer is treated primarily through a multidisciplinary approach that includes surgery, radiation, and systemic medications. Significant efforts have been made over the past ten years to improve each of these therapy strategies, using data from clinical research as well as hints from preclinical and laboratory models. The latter entity has recently been demonstrated to provide highly valuable "from bench to bedside" viewpoints, with substantial potential for finding and utilizing therapeutic targets, while the former is producing significant data through phase I–III trials. This is especially true for radiation science and radiation oncology because this field, which is one of the cornerstones of local treatment, sits at the intersection of several targeted therapy-based tactics. Radiation physics and biology are in fact defined by the growth of active research initiatives, the majority of which—if not all—seek to improve tumour cell killing for an equivalent or diminished impact on normal tissues.[10] In order to improve the therapeutic effectiveness of radiotherapy, comprehensive analyses of cancer cell response to treatment in radiobiology imply to encompass not only conventional parameters like sensitivity levels to radiation but also all mechanisms underlying the response to drugs that may be administered during a radiotherapy treatment. For a variety of solid tumours, including breast malignancies, targeted therapeutics such monoclonal antibodies and small compounds are rigorously studied in clinical settings. Results from treatments based on these non-cytotoxic techniques have been encouraging, but it is evident that further research is needed to fully understand the bio-molecular mechanisms underpinning their capacity to kill cells.

Acknowledgement

The author would like to acknowledge his Department of Biological Chemistry, School of Medicine Research from the University of California for their support during this work.

Conflict of Interest

The author has no known conflicts of interested associated with this paper.

References

  1. Chaber BA, Boral AL, Multani P (1998) Translational Research: Walking the Bridge between Idea and Cure- Seventeenth Bruce F. Cain Memorial Award Lecture.Cancer Res58: 4211–4216.
  2. Indexed at, Google Scholar

  3. Gibbs JB (2000) Mechanism-Based Target Identification and Drug Discovery in Cancer Research.Science 287: 1969–1973.
  4. Indexed at, Google Scholar, Crossref

  5. Saijo N (2002) Translational Study in Cancer.Int Med41: 770–773.
  6. Indexed at, Google Scholar, Crossref

  7. Saijo N, Nishio K, Tamura T (2003) Translational and clinical studies of target-based cancer therapy.Int J Clin Oncol8: 187–192.
  8. Indexed at, Google Scholar, Crossref

  9. Woolf SH (2008) The Meaning of Translational Research and Why It Matters.JAMA 299: 211–213.
  10. Indexed at, Google Scholar, Crossref

  11. Toi M, Takebayashi Y, Chow LW (2005) Translational Research in Breast Cancer.Breast Cancer 12: 86–90.
  12. Indexed at, Google Scholar, Crossref

  13. Mathew JP, Taylor BS, Bader GD, Pyarajan S, Antoniotti M, et al. (2007) From Bytes to Bedside: Data Integration and Computational Biology for Translational Cancer Research.PLoS Comput Biol 3: e12.
  14. Indexed at, Google Scholar, Crossref

  15. Hermiston TW, Kirn DH (2005) Genetically Based Therapeutics for Cancer: Similarities and Contrasts with Traditional Drug Discovery and Development.Mol Ther 11: 496–507.
  16. Indexed at, Google Scholar, Crossref

  17. Beumer HM, Porton WM (1968) Morphologic Effects of Cytotoxic Drugs on Tumor Cells.Diseases Chest 53: 252–256.
  18. Indexed at, Google Scholar, Crossref

  19. Leighton J (1969) Propagation of Cancer: Targets for Future Chemotherapy.Cancer Res29: 2457–2465.
  20. Indexed at, Google Scholar

Citation: Lee WH (2021) The Use of Animal Models in Translational Medicine, the Efficacy of Translational Research in Toxicology, Translational Breast Cancer Research, and Mitochondrial Translational . Int J Drug Dev Res J , Vol.14 No. 6: 959.