GET THE APP

Biomedicine: Development of Organs on Chips
..

Journal of Bioengineering & Biomedical Science

ISSN: 2155-9538

Open Access

Commentary - (2022) Volume 12, Issue 12

Biomedicine: Development of Organs on Chips

Hamid Shirkhanloo*
*Correspondence: Hamid Shirkhanloo, Department of Analytical Chemistry, Institute of Petroleum Industry, Azad University of Tehran, Tehran, Iran, Email:
Department of Analytical Chemistry, Institute of Petroleum Industry, Azad University of Tehran, Tehran, Iran

Received: 28-Nov-2022, Manuscript No. jbbs-23-87932; Editor assigned: 30-Nov-2022, Pre QC No. P-87932; Reviewed: 14-Dec-2022, QC No. Q-87932; Revised: 20-Dec-2022, Manuscript No. R-87932; Published: 28-Dec-2022 , DOI: 10.37421/2155-9538.2022.12.336
Citation: Shirkhanloo, Hamid. “Biomedicine: Development of Organs on Chips.” J Bioengineer & Biomedical Sci 12 (2022): 336.
Copyright: © 2022 Shirkhanloo H. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Description

Organs-on-chips (OOCs), also known as tissue chips or microphysiological frameworks, have emerged as promising in vitro organ models over the past ten years due to their ability to capture organ-level components of human organs and tissues. OOCs enable the production of a close physiological microenvironment by combining a wide range of substances, natural materials, and design science disciplines. OOCs benefit from the advantages of microfluidic innovation, such as low volume, quick reaction, adaptable construction, and precise control of physical and synthetic variables. OOCs can function as in vitro human organ microsystems with improved physiological development and the ability to examine with 2D cell societies and better anticipate human results of various annoyances in relation to creature models thanks to precise control of boundaries such as focus slopes, shear force, cell design, and tissue interface. As a result, OOCs guarantee to overcome any barrier between animal studies and clinical preliminary studies, possibly accelerating the translation of preclinical findings into clinical settings. In this, we present a comprehensive perspective, focusing on the most recent developments in OOCs, the wildernesses of their development, and the most cutting-edge applications, particularly in natural review, disease demonstration, drug disclosure, and preclinical research. We will investigate how to integrate numerous disciplines into OOCs in light of the new accomplishments and anticipate biomedical improvement patterns in the future [1].

In general, using microfabrication and 3D printing, OOCs can be precisely and simultaneously used to recreate numerous aspects of human physiology. The majority of this innovation focuses on the multiplication of the tissue interface, the association of numerous cells at the organ level, and deliberate connections between various organs. The chip can effectively replicate the tissue connection point of human organs by directing relevant cells into bound microchannels or biocompatible platforms. With precise command over outer and inner boundaries like mechanical pressure, dynamic stream, and focus angle, in vitro organ models can demonstrate organ-level association and capability that completely reflects physiological cycles. A progression of parenchymal tissues with useful units, such as the liver, heart, skeletal muscle, and cancers, have been replicated on chip for organic review up until this point, which demonstrates the incredible significance of human physiology. It is important to note that the microfluidic device can be used in exciting studies of early human undeveloped events. The controlled display of human epiblast and amniotic ectoderm from undeveloped cells on a chip by Fu's group has been described in detail. In a similar vein, the group led by Lutolf has developed a cross-genre microprocessor framework for the production of smaller-than-usual stomach tubes made of gastrointestinal undifferentiated cells that are depicted with close physiological spatial plan, which was impossible to achieve with conventional methods. In addition, there have been intense efforts to develop multiorgans-on-chips (MOCs), also known as body-on-a-chip, in order to better comprehend the physiological and obsessive states of the body [2]. The programmable introduction of a dynamic stream into associated organ chambers precisely imitates the blood course in vivo in order to facilitate orderly organ communication. These MOCs microsystems enable the continuous checking of dynamic connections between various organs and constant organ-level responses to improvements at the exterior and interior levels, thereby expanding biomedical applications and potential uses in the future.

The representative for the applications of OOCs frameworks in preclinical measurement, disease detection, drug screening, and organic evaluation. a) A heart-on-a-chip framework that incorporates microphysiological visuality by combining the biohybrid variety hydrogels that underlie it. Considering cell conditions-based primary variety changes and the beating recurrence, the twisting up course of the cantilevered variety hydrogel was initiated by the beating myocardiocytes. A breathing lung-on-a-chip framework for demonstrating human pneumonic edema, which is depicted with impaired obstruction respectability and gas transport, has been reproduced with permission from Reference (b). The significant role that mechanical power plays in the movement of aspiratory edema was demonstrated by this disease model. c) Patient organoids on a chip for creating a microenvironment for pancreatic growth with a variety of cell types and a permeable vascular structure. The microengineered model was also used to test whether chemotherapy drugs were harmful. d) In the drug discovery process, high-throughput MOCs are the stage for continuous checking of connected organs like the human kidney, vascular system, liver, and gastrointestinal tract. The disappointment rate of medication screening may actually be slowed down by this connected MOCs framework. In the future, OOCs microsystems that summarize important organic cycles and reactions in vitro can give new life to numerous applications in disease demonstration, drug disclosure, and preclinical studies. OOCs, in conjunction with patient-specific cells and arising prompted pluripotent immature microorganisms (iPSCs), provide exceptional opportunities to cultivate human in vitro models of diseased organs, enabling in-depth research into fundamental mechanisms underlying disease progression. Numerous OOCs have been developed by various research groups to tailor infection models to individual patient characteristics, such as irritation, apoplexy, cystic fibrosis, Alzheimer's disease, and cancer metastasis, thanks to their superior mimicry of human physiology and pathophysiology. Particularly noteworthy is the use of limited-scope imitations with interconnected multiple organs on chip for the demonstration of muddled diseases. These MOCs frameworks are ideal in vitro models for focusing on complex diseases like those impacted by the safe framework or not a single quality disease. They can restate cooperations between up to ten organs in a single microdevice. MOCs frameworks have been developed by Griffith's group to describe the function of safe cells in ulcerative colitis and other inflammatory diseases, as well as the communication between the metabolic result in the human stomach and Parkinson's disease movement. These infection models certainly have the potential to shed light on the subatomic mechanisms of disease pathogenesis [3].

Over the past ten years, drug researchers have begun to give OOCs more thought because of their small size, cost-viability, isolation, and ability to reproduce in a microenvironment similar to that of a living organism. The drug industry is currently facing enormous challenges that can be attributed to the rising cost of normal cell and creature models and their limited predictive power. There are roughly two areas where OOCs innovation can significantly advance the pipeline for medication improvement: boosting the medication success rate and individualizing the medication. First and foremost, microengineered OOCs can serve as new testing models for accurate expectations of medication adequacy and security, reducing both the cost and the rate of medication failure. This is difficult for standard 2D cell and creature models because these models are unable to accurately represent human physiology due to differences in structure and species. With a library of new medications, the next direct step is a high-throughput screening measure. As a result, efforts have been made to develop OOCs models with high throughput rates and reproducibility for identifying and breaking down drug reactions. Additionally, these chip devices are adaptable to multisensors, enabling extremely dynamic and delicate discovery and detachment on a single cell or tissue scale. Due to the small fluid volumes created by restricted microchannels and hydrogel drops on chips, drug screening in a microfluidic culture framework with improved paracrine and autocrine motioning may also be significantly improved. In addition, the inherent advantages of OOCs, such as their simple activity, precise reagent control, low dissipation, and optical simplicity for imaging and investigation [4], can increase the success rate of medication tests.

According to quiet's own highlights, customized medication has always been regarded as tailored preventive measures and focused, advanced treatments. Customized OOCs frameworks can be created using patient samples or cells to show how each patient responds to the right dosage and medication. Amazingly, advances in iPSC research have the potential to develop tissue or organ models that are specific to patients and diseases. Customized drug testing has been accounted for by a variety of human iPSC-based OOCs models, including those for Parkinson's disease, Duchenne muscle dystrophy, schizophrenia, nonalcoholic steatohepatitis, type 1 diabetes, and polycystic kidney disease. In addition, the capacity of human immature microorganisms' inferred organoid innovation is staggering. Organoids are flexible tools for in vitro drug disclosure because they are three-dimensional miniature tissues that feature in vivo-like cell parts, design, and capabilities. Patient-explicit organoids with hereditary mutations can be created with the help of reimagined iPSCs derived from patient substantial cells [5]. These organoids provide customized infection models for precise medication. Even with the advancements in organoid technology, it is still challenging to develop drug testing frameworks based on organoids because the current organoids are still quite young when compared to local organs even after long-term culture. On the other hand, malignant growth organoids derived from patient disease tissues may be able to consistently maintain the morphological and inherited characteristics of the initial cancers and may even be able to serve as a reliable option for restorative evaluation of individual patients. Several striking studies have been conducted to identify persistently determined digestive, ovarian, pancreatic, prostate, kidney, and cellular breakdown in lungs organoids for extensive drug screening and the expectation of resistance to chemotherapy and radiation. These agent studies indicate that the OOCs microengineering strategy may open new doors to more powerful preclinical studies of human medication reactions.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Huh, Dongeun, Daniel C. Leslie, Benjamin D. Matthews and Jacob P. Fraser, et al. "A human disease model of drug toxicity–induced pulmonary edema in a lung-on-a-chip microdevice." Sci Transl Med 4 (2012): 159ra147-159ra147.
  2. Google Scholar, Crossref, Indexed at

  3. Lai, Benjamin Fook Lun, Rick Xing Ze Lu, Yangshuo Hu and Locke Davenport Huyer, et al. "Recapitulating pancreatic tumor microenvironment through synergistic use of patient organoids and organ‐on‐a‐chip vasculature.” Adv Funct Mater 30 (2020): 2000545.
  4. Google Scholar, Crossref, Indexed at

  5. Azizgolshani, H., J.R. Coppeta, E.M. Vedula and E.E. Marr, et al. "High-throughput organ-on-chip platform with integrated programmable fluid flow and real-time sensing for complex tissue models in drug development workflows." Lab Chip 21 (2021): 1454-1474.
  6. Google Scholar, Crossref, Indexed at

  7. Zhang, Boyang, Anastasia Korolj, Benjamin Fook Lun Lai and Milica Radisic. "Advances in organ-on-a-chip engineering." Nat Rev Mater 3 (2018): 257-278.
  8. Google Scholar, Crossref, Indexed at

  9. Fu, Fanfan, Luoran Shang, Zhuoyue Chen, Yunru Yu and Yuanjin Zhao. "Bioinspired living structural color hydrogels." Sci Robot 3 (2018): eaar8580.
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 307

Journal of Bioengineering & Biomedical Science received 307 citations as per Google Scholar report

Journal of Bioengineering & Biomedical Science peer review process verified at publons

Indexed In

arrow_upward arrow_upward