Summary

Robust Mitochondrial Isolation from Rodent Cardiac Tissue

Published: August 23, 2024
doi:

Summary

Bioenergetic and metabolomic studies on mitochondria have revealed their multifaceted role in many diseases, but the isolation methods for these organelles vary. The method detailed here is capable of purifying high-quality mitochondria from multiple tissue sources. Quality is determined by respiratory control ratios and other metrics assessed with high-resolution respirometry.

Abstract

Mitochondrial isolation has been practiced for decades, following procedures established by pioneers in the fields of molecular biology and biochemistry to study metabolic impairments and disease. Consistent mitochondrial quality is necessary to properly investigate mitochondrial physiology and bioenergetics; however, many different published isolation methods are available for researchers. Although different experimental strategies require different isolation methods, the basic principles and procedures are similar. This protocol details a method capable of extracting well-coupled mitochondria from a variety of tissue sources, including small animals and cells. The steps outlined include organ dissection, mitochondrial purification, protein quantification, and various quality control checks. The primary quality control metric used to identify high-quality mitochondria is the respiratory control ratio (RCR). The RCR is the ratio of the respiratory rate during oxidative phosphorylation to the rate in the absence of ADP. Alternative metrics are discussed. While high RCR values relative to their tissue source are obtained using this protocol, several steps can be optimized to suit the individual needs of researchers. This procedure is robust and has consistently resulted in isolated mitochondria with above-average RCR values across animal models and tissue sources.

Introduction

Mitochondria are subcellular organelles that establish cytoplasmic energetic conditions optimized for specific cell functions. While cellular, tissue, and organism-level studies can provide insights into mitochondrial function, isolating the organelles offers a level of experimental control not possible otherwise. Mitochondrial isolations have been performed since the 1940s, allowing mechanistic studies of metabolism and respiration across a variety of cells and tissues1,2. The historical relevance of mitochondria is also well-documented3. As the main producers of ATP, mitochondria play many key roles that are vital for optimal cellular and organ function4. Within the mitochondrial matrix, substrates are oxidized by the TCA cycle, producing reducing equivalents and mobile electron carriers such as NADH and UQH25,6. Cytochrome C is the third main mobile electron carrier in the mitochondrial biochemical reaction network7. These molecules are then oxidized by the transmembrane complexes of the electron transport system (ETS) embedded in the inner mitochondrial membrane8. Redox reactions of the ETS are coupled with proton translocation from the matrix to the intermembrane space. These processes establish an electrochemical proton gradient that is used to phosphorylate ADP with Pi by F1F0 ATP synthase to produce ATP9,10. The individual processes that occur at each complex can be explored with high-resolution respirometry using Clark-type electrodes or microplate oxygen consumption assays11,12. Additionally, disease models and treatments using isolated mitochondria can determine the impact or importance of mitochondrial function in the progression of certain pathologies. This has proven fruitful in the field of cardiology, where alterations in fuel and substrate delivery have been used to elucidate how mitochondrial dysfunction influences heart failure13,14,15,16. Mitochondria are also known to impact the development of other disease states such as diabetes, cancer, obesity, neurological disorders, and myopathies17,18. Therefore, the use of isolated or purified mitochondria enables mechanistic investigations of oxidative metabolism and ATP production in the source tissue.

There is no shortage of mitochondrial isolation protocols due to their importance in bioenergetic research. Additionally, highly specific methods tailored to subpopulations of mitochondria within tissues and cells can be found19,20. The basic procedural steps are similar between isolation methods, but variations can be made to buffer composition, homogenization steps, and centrifugation spins to improve the amount and quality of mitochondria. Changes to these aspects are based on the metabolic demand of the tissue, overall organ function, mitochondrial density, and other factors. In tissues such as the liver and skeletal muscle, handheld homogenizers are used to preserve mitochondrial integrity and limit damage to the mitochondrial membranes21. However, when isolating from kidneys, some protocols suggest using manually driven homogenization or commercial kits to yield better results22. Although both methods yield functional mitochondria, the quality of the organelles can become compromised by the additional time it takes to complete isolations using these protocols. Centrifugation is also vital to the extraction of mitochondrial protein, as it separates cellular components such as nuclei and other organelles from mitochondria23. During the isolation process, it is debated whether differential or density-based centrifugation should be implemented to obtain purer isolates24. While density centrifugation can separate mitochondria from organelles of similar specific gravity, such as peroxisomes, it is not well-established if mitochondria from these methods better represent in situ organelle function compared to mitochondria isolated using differential centrifugation. In the field of mitochondrial physiology, density-based centrifugation is preferred and can be easily altered to increase isolate purity. Whether changes to g-forces, centrifugation duration, and the number of centrifugation spins are incorporated should be considered before experimentation due to their influence on mitochondrial purification. Furthermore, mitochondrial resuspension, arguably the most crucial step during isolation, differs greatly between studies, with the use of scraping, vortex-based mixing, or homogenization23,25,26. Mechanical resuspension of these types can be too abrasive and impair the membrane integrity of mitochondria. For this reason, gentle washing should be performed to correct this. Despite the plethora of modulations and suggested methodologies, there are fewer comprehensive protocols with high reproducibility and adaptability for rodent models.

The methods described herein outline a detailed, robust, and highly reproducible protocol that yields purified, well-coupled mitochondria from small animal cardiac tissue. As demonstrated, this method can be easily adapted to accommodate the specific needs of each experiment and/or laboratory environment for isolating mitochondria from kidneys, liver, and cultured cells. Further modifications can be made to isolate mitochondria from tissues and other animals not listed here. Buffer recipes used for all isolations are provided and can be modified if needed. Similar to other published protocols, motorized homogenization, and differential centrifugation are implemented; however, adjustments are made to both the shearing time and the force at which the samples are centrifuged to consistently deliver high-quality mitochondrial isolates, depending on the isolation source. Notably, this protocol differs from others by using gentle washing via pipetting to resuspend pelleted mitochondria, which helps preserve mitochondrial membrane integrity and maintain the overall functionality of the organelles. Mitochondrial protein is quantified either by total protein determination or by measuring citrate synthase activity. The utility and broad applicability of this isolation method are further supported by the values of respiratory control ratios (RCR) achieved across various organisms and tissue sources.

Protocol

The use and treatment of all vertebrate animals were performed in accordance with approved protocols reviewed and accepted by the Institutional Animal Care and Use Committee (IACUC) at Michigan State University. This protocol was designed using both male and female Hartley albino guinea pigs and Sprague Dawley (SD) rats. For the isolation of cardiac mitochondria from guinea pigs, animals were sacrificed at ages between 4-6 weeks (300-450 g). Cardiac mitochondria from SD rats of both sexes were obtained between the ages o…

Representative Results

Upon completion of mitochondrial isolation, the quality and functionality of the isolates should be tested via quantifying rates of oxygen consumption (JO2) using high-resolution respirometry. To do so, mitochondrial stocks were diluted to 40 mg/mL to allow for working concentrations of 0.1 mg/mL in 2 mL of RB for all respirometry assays using isolated cardiac mitochondria. Respiration was fueled by 5 mM sodium pyruvate and 1 mM L-malate in the presence of 1 mM EGTA, a calcium chelator, and was allowe…

Discussion

Adhering to the methods concisely described in this protocol will ensure the isolation of well-coupled mitochondria from the cardiac tissue of small rodents, in addition to other tissue types and sources. Overall, the process should take a total of 3-3.5 h, during which all animal tissue, samples, and isolates should remain on ice at 4 °C as much as possible to limit degradation. This procedure is robust and can be altered in several ways to better fit experimental goals and models utilized. One modulation that can …

Offenlegungen

The authors have nothing to disclose.

Acknowledgements

We would like to acknowledge Daniel A. Beard and Kalyan C. Vinnakota for their foundational contributions to this protocol. This work was funded by NSF CAREER grant MCB-2237117.

Materials

1.7 mL microcentrifuge tubes
10 mL glass beaker For organ disection and mincing
50 mL centrifuge tubes Centrifugation
Adenosine 5'-diphosphate monopotassium salt dihydrate Sigma  A5285 Respirometry assays
BSA Protein Assay Kit Thermo Scentific PI23225 Mitochondrial protein quantification
Dextrose Sigma  DX0145 For buffer (CB)
Ethylene glycol-bis(2-amino-ethylether)-N,N,N',N'-tetraacetic acid  Sigma  E4378 For buffers (CB and IB) and respirometry assays
Glass cannula Radnoti Guinea pig and rat heart perfusion
Heparin sodium porcine mucosa Sigma  SRE0027-250KU Animal IP injection
High-resolution respirometer Clark-type electrode; oxygraph with 2 mL chambers
Induction chamber
Isoflurane Sigma  792632 Anesthetic 
L-malic acid Sigma  02288-50G Respirometry assays
Magnesium chloride hexahydrate Sigma  M9272 For buffer (RB)
Mannitol Sigma  MX0214 For buffer (IB)
Microliter syringes Sizes ranging from 5–50 µL
Microplate reader Must be able to incubate at 37 °C 
MOPS Sigma  475898 For all buffers
O2 tank
OMNI THQ Homogenizer OMNI International  12-500 Similar rotor stator homogenizers will work
pipettes Volumes of  2–20 µL; 20–200 µL; 200–1000 µL
Potassium chloride Sigma  P3911 For buffers (RB and CB)
Potassium phosphate dibasic Sigma  795496 For buffers (IB and RB)
Protease from Bacillus licheniformis Sigma  P5459
Sodium chloride Sigma  S9888 For buffer (CB)
Sodium pyruvate Fisher bioreagents BP356-100 Respirometry assays
Sucrose Sigma  8510-OP For buffer (IB)
Surgical dissection kit Depends on animal and tissue source
Tabletop centrifuge Must cool to 4 °C 

Referenzen

  1. Hogeboom, G. H., Schneider, W. C., Pallade, G. E. Cytochemical studies of mammalian tissues; isolation of intact mitochondria from rat liver; some biochemical properties of mitochondria and submicroscopic particulate material. J Biol Chem. 172 (2), 619-635 (1948).
  2. Claude, A. Fractionation of mammalian liver cells by differential centrifugation: Ii. Experimental procedures and results. J Exp Med. 84 (1), 61-89 (1946).
  3. Ernster, L. Schatz, G. Mitochondria: A historical review. J Cell Biol. 91 (3 Pt 2), 227s-255s (1981).
  4. Martinez-Reyes, I. Chandel, N. S. Mitochondrial tca cycle metabolites control physiology and disease. Nat Commun. 11 (1), 102 (2020).
  5. Vercellino, I. Sazanov, L. A. The assembly, regulation, and function of the mitochondrial respiratory chain. Nat Rev Mol Cell Biol. 23 (2), 141-161 (2022).
  6. Fernie, A. R., Carrari, F., Sweetlove, L. J. Respiratory metabolism: Glycolysis, the TCA cycle and mitochondrial electron transport. Curr Opin Plant Biol. 7 (3), 254-261 (2004).
  7. Kalpage, H. A. et al. Cytochrome C phosphorylation: Control of mitochondrial electron transport chain flux and apoptosis. Int J Biochem Cell Biol. 121, 105704 (2020).
  8. Sousa, J. S., D'imprima, E., Vonck, J. Mitochondrial respiratory chain complexes. Subcell Biochem. 87, 167-227 (2018).
  9. Mitchell, P. Moyle, J. Chemiosmotic hypothesis of oxidative phosphorylation. Nature. 213 (5072), 137-139 (1967).
  10. Chance, B. Williams, G. R. Respiratory enzymes in oxidative phosphorylation. I. Kinetics of oxygen utilization. J Biol Chem. 217 (1), 383-393 (1955).
  11. Li, Z. Graham, B. H. Measurement of mitochondrial oxygen consumption using a Clark electrode. Methods Mol Biol. 837, 63-72 (2012).
  12. Rogers, G. W. et al. High throughput microplate respiratory measurements using minimal quantities of isolated mitochondria. PLoS One. 6 (7), e21746 (2011).
  13. Bisaccia, G., Ricci, F., Gallina, S., Di Baldassarre, A., Ghinassi, B. Mitochondrial dysfunction and heart disease: Critical appraisal of an overlooked association. Int J Mol Sci. 22 (2), 614 (2021).
  14. Zhou, B. Tian, R. Mitochondrial dysfunction in pathophysiology of heart failure. J Clin Invest. 128 (9), 3716-3726 (2018).
  15. Holzem, K. M. et al. Mitochondrial structure and function are not different between nonfailing donor and end-stage failing human hearts. FASEB J. 30 (8), 2698-2707 (2016).
  16. Cordero-Reyes, A. M. et al. Freshly isolated mitochondria from failing human hearts exhibit preserved respiratory function. J Mol Cell Cardiol. 68, 98-105 (2014).
  17. Norat, P. et al. Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen Med. 5 (1), 22 (2020).
  18. Diaz-Vegas, A. et al. Is mitochondrial dysfunction a common root of noncommunicable chronic diseases? Endocr Rev. 41 (3), bnaa005 (2020).
  19. Lai, N. et al. Isolation of mitochondrial subpopulations from skeletal muscle: Optimizing recovery and preserving integrity. Acta Physiol (Oxf). 225 (2), e13182 (2019).
  20. Holmuhamedov, E. L., Oberlin, A., Short, K., Terzic, A., Jahangir, A. Cardiac subsarcolemmal and interfibrillar mitochondria display distinct responsiveness to protection by diazoxide. PLoS One. 7 (9), e44667 (2012).
  21. Frezza, C., Cipolat, S., Scorrano, L. Organelle isolation: Functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat Protoc. 2 (2), 287-295 (2007).
  22. Micakovic, T. et al. Isolation of pure mitochondria from rat kidneys and western blot of mitochondrial respiratory chain complexes. Bio Protoc. 9 (19), e3379 (2019).
  23. Liao, P. C., Bergamini, C., Fato, R., Pon, L. A., Pallotti, F. Isolation of mitochondria from cells and tissues. Methods Cell Biol. 155, 3-31 (2020).
  24. Hubbard, W. B. et al. Fractionated mitochondrial magnetic separation for isolation of synaptic mitochondria from brain tissue. Sci Rep. 9 (1), 9656 (2019).
  25. Zhou, D. et al. Protocol for mitochondrial isolation and sub-cellular localization assay for mitochondrial proteins. STAR Protoc. 4 (1), 102088 (2023).
  26. Hernandez-Camacho, J. D. et al. Isolation of mitochondria from mouse skeletal muscle for respirometric assays. J Vis Exp. 180, e63336 (2022).
  27. Farrugia, G. E. et al. A protocol for rapid and parallel isolation of myocytes and non-myocytes from multiple mouse hearts. STAR Protoc. 2 (4), 100866 (2021).
  28. Wollenman, L. C., Vander Ploeg, M. R., Miller, M. L., Zhang, Y., Bazil, J. N. The effect of respiration buffer composition on mitochondrial metabolism and function. PLoS One. 12 (11), e0187523 (2017).
  29. Vinnakota, K. C. et al. Open-loop control of oxidative phosphorylation in skeletal and cardiac muscle mitochondria by ca(2.). Biophys J. 110 (4), 954-961 (2016).
  30. Vinnakota, K. C., Bazil, J. N., Van Den Bergh, F., Wiseman, R. W., Beard, D. A. Feedback regulation and time hierarchy of oxidative phosphorylation in cardiac mitochondria. Biophys J. 110 (4), 972-980 (2016).
  31. Warnock, L. B. Huang, D. in Statpearls., Treasure Island (FL) ineligible companies. Disclosure: Davis Huang declares no relevant financial relationships with ineligible companies. (2024).
  32. Sercel, A. J. et al. Generating stable isolated mitochondrial recipient clones in mammalian cells using mitopunch mitochondrial transfer. STAR Protoc. 2 (4), 100850 (2021).
  33. Lampl, T., Crum, J. A., Davis, T. A., Milligan, C., Del Gaizo Moore, V. Isolation and functional analysis of mitochondria from cultured cells and mouse tissue. J Vis Exp. 97, e52076 (2015).
  34. Sobolewski, P., Kandel, J., Eckmann, D. M. Air bubble contact with endothelial cells causes a calcium-independent loss in mitochondrial membrane potential. PLoS One. 7 (10), e47254 (2012).
  35. Bradford, M. M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72, 248-254 (1976).
  36. Wei, P., Liu, Q., Xue, W., Wang, J. A colorimetric assay of citrate synthase activity in drosophila melanogaster. J Vis Exp. 155, e59454 (2020).
  37. Tomar, N. et al. Substrate-dependent differential regulation of mitochondrial bioenergetics in the heart and kidney cortex and outer medulla. Biochim Biophys Acta Bioenerg. 1863 (2), 148518 (2022).
  38. Ter Veld, F., Jeneson, J. A., Nicolay, K. Mitochondrial affinity for ADP is twofold lower in creatine kinase knock-out muscles. Possible role in rescuing cellular energy homeostasis. FEBS J. 272 (4), 956-965 (2005).
  39. Gouspillou, G. et al. Accurate determination of the oxidative phosphorylation affinity for ADP in isolated mitochondria. PLoS One. 6 (6), e20709 (2011).
  40. Strubbe-Rivera, J. O. et al. The mitochondrial permeability transition phenomenon elucidated by cryo-em reveals the genuine impact of calcium overload on mitochondrial structure and function. Sci Rep. 11 (1), 1037 (2021).
  41. Zhou, Z. et al. Diverse functions of cytochrome C in cell death and disease. Cell Death Differ. 31 (4), 387-404 (2024).
  42. Certo, M. et al. Mitochondria primed by death signals determine cellular addiction to antiapoptotic bcl-2 family members. Cancer Cell. 9 (5), 351-365 (2006).
  43. Bose, S., French, S., Evans, F. J., Joubert, F., Balaban, R. S. Metabolic network control of oxidative phosphorylation: Multiple roles of inorganic phosphate. J Biol Chem. 278 (40), 39155-39165 (2003).
  44. Motawe, Z. Y., Abdelmaboud, S. S., Breslin, J. W. Evaluation of glycolysis and mitochondrial function in endothelial cells using the seahorse analyzer. Methods Mol Biol. 2711, 241-256 (2024).
  45. Will, Y., Hynes, J., Ogurtsov, V. I., Papkovsky, D. B. Analysis of mitochondrial function using phosphorescent oxygen-sensitive probes. Nat Protoc. 1 (6), 2563-2572 (2006).
  46. Huerta, B. et al. Sea lamprey cardiac mitochondrial bioenergetics after exposure to TFM and its metabolites. Aquat Toxicol. 219, 105380 (2020).
  47. Malyala, S., Zhang, Y., Strubbe, J. O., Bazil, J. N. Calcium phosphate precipitation inhibits mitochondrial energy metabolism. PLoS Comput Biol. 15 (1), e1006719 (2019).
This article has been published
Video Coming Soon
Keep me updated:

.

Diesen Artikel zitieren
Vadovsky, A. C., Quinn, M., Xia, T., Levitsky, Y., Bazil, J. N. Robust Mitochondrial Isolation from Rodent Cardiac Tissue. J. Vis. Exp. (210), e67093, doi:10.3791/67093 (2024).

View Video