1
|
Kusari F, Backova L, Panek D, Benda A, Trachtulec Z. Label-free metabolic fingerprinting of motile mammalian spermatozoa with subcellular resolution. BMC Biol 2025; 23:85. [PMID: 40128804 PMCID: PMC11934609 DOI: 10.1186/s12915-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Sperm metabolic pathways that generate energy for motility are compartmentalized within the flagellum. Dysfunctions in metabolic compartments, namely mitochondrial respiration and glycolysis, can compromise motility and male fertility. Studying these compartments is thus required for fertility treatment. However, it is very challenging to capture images of metabolic compartments in motile spermatozoa because the fast beating of the flagellum introduces motion blur. Therefore, most approaches immobilize spermatozoa prior to imaging. RESULTS Our findings indicate that immobilizing sperm alters their metabolic profile, highlighting the necessity for measuring metabolism in spermatozoa during movement. We achieved this by encapsulating mouse epididymis in a hydrogel followed by two-photon fluorescence lifetime imaging microscopy for imaging motile sperm in situ. The autofluorescence of endogenous metabolites-FAD, NADH, and NADPH-enabled us to visualize sperm metabolic compartments without staining. We trained machine learning for automated image segmentation and generated metabolic fingerprints using object-based phasor analysis. We show that metabolic fingerprints of spermatozoa and the mitochondrial compartment (1) can distinguish individual males by genetic background, age, or fecundity status, (2) correlate with fertility, and (3) change with age likely due to increased oxidative metabolism. CONCLUSIONS Our approach eliminates the need for sperm immobilization and labeling and captures the native state of sperm metabolism. This technique could be adapted for metabolism-based sperm selection for assisted reproduction.
Collapse
Affiliation(s)
- Fitore Kusari
- Laboratory of Germ Cell Development, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
- Present address: Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Lenka Backova
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
- Present addresses: Instituto Biofisika CSIC, UPV/EHU, Leioa, Spain
- Department of Computer Science and Artificial Intelligence, University of Basque Country UPV/EHU, San Sebastián, Spain
| | - Dalibor Panek
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ales Benda
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Zdenek Trachtulec
- Laboratory of Germ Cell Development, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
Rao X, Liu W. A Guide to Metabolic Network Modeling for Plant Biology. PLANTS (BASEL, SWITZERLAND) 2025; 14:484. [PMID: 39943046 PMCID: PMC11820892 DOI: 10.3390/plants14030484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025]
Abstract
Plants produce a diverse array of compounds that play crucial roles in growth, in development, and in responses to abiotic and biotic stresses. Understanding the fluxes within metabolic pathways is essential for guiding strategies aimed at directing metabolism for crop improvement and the plant natural product industry. Over the past decade, metabolic network modeling has emerged as a predominant tool for the integration, quantification, and prediction of the spatial and temporal distribution of metabolic flows. In this review, we present the primary methods for constructing mathematical models of metabolic systems and highlight recent achievements in plant metabolism using metabolic modeling. Furthermore, we discuss current challenges in applying network flux analysis in plants and explore the potential use of machine learning technologies in plant metabolic modeling. The practical application of mathematical modeling is expected to provide significant insights into the structure and regulation of plant metabolic networks.
Collapse
Affiliation(s)
- Xiaolan Rao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Wei Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, China
| |
Collapse
|
3
|
Lin P, Zhang S, Komatsubara F, Konishi H, Nakata E, Morii T. Artificial Compartments Encapsulating Enzymatic Reactions: Towards the Construction of Artificial Organelles. Chempluschem 2025; 90:e202400483. [PMID: 39351818 DOI: 10.1002/cplu.202400483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Cells have used compartmentalization to implement complex biological processes involving thousands of enzyme cascade reactions. Enzymes are spatially organized into the cellular compartments to carry out specific and efficient reactions in a spatiotemporally controlled manner. These compartments are divided into membrane-bound and membraneless organelles. Mimicking such cellular compartment systems has been a challenge for years. A variety of artificial scaffolds, including liposomes, polymersomes, proteins, nucleic acids, or hybrid materials have been used to construct artificial membrane-bound or membraneless compartments. These artificial compartments may have great potential for applications in biosynthesis, drug delivery, diagnosis and therapeutics, among others. This review first summarizes the typical examples of cellular compartments. In particular, the recent studies on cellular membraneless organelles (biomolecular condensates) are reviewed. We then summarize the recent advances in the construction of artificial compartments using engineered platforms. Finally, we provide our insights into the construction of biomimetic systems and the applications of these systems. This review article provides a timely summary of the relevant perspectives for the future development of artificial compartments, the building blocks for the construction of artificial organelles or cells.
Collapse
Affiliation(s)
- Peng Lin
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Shiwei Zhang
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Futa Komatsubara
- Graduate School of Energy Science, Kyoto University, Yoshida-hommachi, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hiroaki Konishi
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Eiji Nakata
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
| | - Takashi Morii
- Institute of Advanced Energy, Kyoto University, Uji-shi, Kyoto, 611-0011, Japan
- Department of Health and Nutrition, Kyoto Koka Women's University, Ukyo-ku, Kyoto, 615-0882, Japan
| |
Collapse
|
4
|
Wu H, Qin B, Yang G, Ji P, Gao Y, Zhang L, Wang B, Liu G. The Protective Effects of Melatonin on Hainan Black Goats Under Heat Stress: Understanding Its Actions and Mechanisms. Antioxidants (Basel) 2025; 14:44. [PMID: 39857379 PMCID: PMC11760882 DOI: 10.3390/antiox14010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
As the global climate changes, high temperatures will cause heat stress, which significantly affects the productive efficiency of livestock. Currently, there is a lack of efficient methods to use in targeting this issue. In this study, we report that melatonin supplementation may represent an alternative method to reduce the negative impact of heat stress on livestock, particularly in Hainan black goats. Our results show that melatonin treatment increased the average daily gain of Hainan black goats that were exposed to constantly high temperatures for two months compared to controls. Our mechanistic exploration revealed that melatonin treatment not only reduced the oxidative stress and inflammatory reaction caused by heat stress but also improved goats' metabolic capacity, promoting their growth and development. More importantly, for the first time, we observed that melatonin treatment modified the abundance of the intestinal microflora, altering the metabolism of the goats, which further improved their tolerance to constant heat stress.
Collapse
Affiliation(s)
- Hao Wu
- Sany Institute of China Agricultural University, Sanya 572025, China; (H.W.); (B.Q.); (G.Y.)
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Baochun Qin
- Sany Institute of China Agricultural University, Sanya 572025, China; (H.W.); (B.Q.); (G.Y.)
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Guang Yang
- Sany Institute of China Agricultural University, Sanya 572025, China; (H.W.); (B.Q.); (G.Y.)
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Pengyun Ji
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Yu Gao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Lu Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Bingyuan Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| | - Guoshi Liu
- Sany Institute of China Agricultural University, Sanya 572025, China; (H.W.); (B.Q.); (G.Y.)
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.J.); (Y.G.); (L.Z.)
| |
Collapse
|
5
|
Norden PR, Wedan RJ, Longenecker JZ, Preston SEJ, Graber N, Pentecost OA, Canfield M, McLaughlin E, Nowinski SM. Mitochondrial Phosphopantetheinylation is Required for Oxidative Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.592977. [PMID: 38766035 PMCID: PMC11100772 DOI: 10.1101/2024.05.09.592977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
4'-phosphopantetheinyl (4'PP) groups are essential co-factors added to target proteins by p hospho p antetheinyl transferase (PPTase) enzymes. Although mitochondrial 4'PP-modified proteins have been described for decades, a mitochondrially-localized PPTase has never been found in mammals. We discovered that the cytoplasmic PPTase a mino a dipate s emialdehyde d ehydrogenase p hospho p antetheinyl t ransferase (AASDHPPT) is required for mitochondrial respiration and oxidative metabolism. Loss of AASDHPPT results in failed 4'-PP modification of the mitochondrial acyl carrier protein and blunted activity of the mitochondrial fatty acid synthesis (mtFAS) pathway. We found that in addition to its cytoplasmic localization, AASDHPPT localizes to the mitochondrial matrix via an N-terminal mitochondrial targeting sequence contained within the first 13 amino acids of the protein. Our data show that this novel mitochondrial localization of AASDHPPT is required to support mtFAS activity and oxidative function. We further identify two variants of uncertain significance in AASDHPPT that are likely pathogenic in humans due to loss of mtFAS activity.
Collapse
|
6
|
Xiong F, Wei J, Zhou Y, Shao Y, Liu J, Chen F. Exploring the Subcellular Localization of Monascus Pigments Biosynthases: Preliminary Unraveling of the Compartmentalization Mechanism. J Fungi (Basel) 2024; 10:375. [PMID: 38921362 PMCID: PMC11205011 DOI: 10.3390/jof10060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Monascus pigments (MPs), a class of secondary metabolites produced by Monascus spp., can be classified into yellow, orange, and red MPs according to their differences in the wavelength of the maximum absorption. However, the biosynthetic sequence and cellular biosynthesis mechanism of different MPs components are still not yet completely clear in Monascus spp. In this study, the subcellular localization of five MPs synthases was investigated using fluorescent protein fusion expression. The results revealed that the proteins encoded by the MPs biosynthetic gene cluster were compartmentalized in various subcellular locations, including the mitochondrial polyketide synthase MrPigA, cytosolic enzymes consisting of the ketoreductase MrPigC, the oxidoreductase MrPigE, and the monooxygenase MrPigN, and the cell-wall-bound oxidoreductase MrPigF. Moreover, the correct localization of MrPigF to the cell wall was crucial for the synthesis of orange MPs. Lastly, we discussed the compartmentalized biosynthetic pathway of MPs. This study will not only be helpful in clarifying the biosynthetic sequence and biosynthesis mechanism of different MPs but also provides new insights into the cellular biosynthesis of secondary metabolites in filamentous fungi.
Collapse
Affiliation(s)
- Fei Xiong
- National Key Laboratory of Agricultural Microbiology, Hubei International Scientific and Technological Cooperation Base of Traditional Fermented Foods, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingyi Wei
- National Key Laboratory of Agricultural Microbiology, Hubei International Scientific and Technological Cooperation Base of Traditional Fermented Foods, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Youxiang Zhou
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-Products, Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Yanchun Shao
- National Key Laboratory of Agricultural Microbiology, Hubei International Scientific and Technological Cooperation Base of Traditional Fermented Foods, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiao Liu
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-Products, Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Fusheng Chen
- National Key Laboratory of Agricultural Microbiology, Hubei International Scientific and Technological Cooperation Base of Traditional Fermented Foods, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
7
|
Parlakgül G, Pang S, Artico LL, Min N, Cagampan E, Villa R, Goncalves RLS, Lee GY, Xu CS, Hotamışlıgil GS, Arruda AP. Spatial mapping of hepatic ER and mitochondria architecture reveals zonated remodeling in fasting and obesity. Nat Commun 2024; 15:3982. [PMID: 38729945 PMCID: PMC11087507 DOI: 10.1038/s41467-024-48272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
The hepatocytes within the liver present an immense capacity to adapt to changes in nutrient availability. Here, by using high resolution volume electron microscopy, we map how hepatic subcellular spatial organization is regulated during nutritional fluctuations and as a function of liver zonation. We identify that fasting leads to remodeling of endoplasmic reticulum (ER) architecture in hepatocytes, characterized by the induction of single rough ER sheet around the mitochondria, which becomes larger and flatter. These alterations are enriched in periportal and mid-lobular hepatocytes but not in pericentral hepatocytes. Gain- and loss-of-function in vivo models demonstrate that the Ribosome receptor binding protein1 (RRBP1) is required to enable fasting-induced ER sheet-mitochondria interactions and to regulate hepatic fatty acid oxidation. Endogenous RRBP1 is enriched around periportal and mid-lobular regions of the liver. In obesity, ER-mitochondria interactions are distinct and fasting fails to induce rough ER sheet-mitochondrion interactions. These findings illustrate the importance of a regulated molecular architecture for hepatocyte metabolic flexibility.
Collapse
Affiliation(s)
- Güneş Parlakgül
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Song Pang
- HHMI Janelia Research Campus, Ashburn, VA, USA
- Yale School of Medicine, New Haven, CT, USA
| | - Leonardo L Artico
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Nina Min
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Erika Cagampan
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reyna Villa
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Renata L S Goncalves
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Grace Yankun Lee
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - C Shan Xu
- HHMI Janelia Research Campus, Ashburn, VA, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
8
|
Li G, Huang Y, Zhao L, Yang B, Guo J, Hu J, Wang J, Wang H, Liu B, Zhang A, Sun F, Luo Q. Targeting and Microenvironment-Activated Nanoreactor for Diabetic Chronic Wound Healing via Multienzyme Cascade Reactions. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6315-6326. [PMID: 38277498 DOI: 10.1021/acsami.3c12427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
The development of cell-like nanoreactors with the ability to initiate biocatalytic cascades under special conditions holds tremendous potential for therapeutic applications. Herein, conformationally gated nanoreactors that respond to the acidic microenvironment of infected diabetic wounds were developed by cucur[8]bituril (CB[8])-based supramolecular assembly. The bioinspired nanoreactors exhibit not only self-regulated permeability and selectivity to control internal enzyme activities by substance exchange but also distinct binding specificities toward Gram-positive and Gram-negative bacteria via noncovalent modification with different ligands. The encapsulation of glucose oxidase (GOx), Fe3O4 nanozyme, and l-arginine (l-Arg) into the nanocarriers enables intelligent activation of multienzyme cascade reactions upon glucose (Glu) uptake to produce gluconic acid (GA) and hydrogen peroxide (H2O2), which is further converted into highly toxic hydroxyl radicals (·OH) for selective antibacterial activity. Moreover, acidic H2O2 promotes the oxidization of l-Arg, leading to the release of nitric oxide (NO). Consequently, this nanoreactor provides a multifunctional and synergistic platform for diabetic chronic wound healing by combining enzyme dynamic therapy with NO gas therapy to combat bacterial infections and inflammation under high blood Glu levels.
Collapse
Affiliation(s)
- Ge Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yibing Huang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Linlu Zhao
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Bo Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiale Guo
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Juntao Hu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jinli Wang
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Hui Wang
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Bin Liu
- Jilin Province Product Quality Supervision and Inspection Institute, No.2699 Yiju Road, Changchun 130103, China
| | - Aiguo Zhang
- Jilin Province Product Quality Supervision and Inspection Institute, No.2699 Yiju Road, Changchun 130103, China
| | - Fengying Sun
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Quan Luo
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
9
|
Alam S, Doherty E, Ortega-Prieto P, Arizanova J, Fets L. Membrane transporters in cell physiology, cancer metabolism and drug response. Dis Model Mech 2023; 16:dmm050404. [PMID: 38037877 PMCID: PMC10695176 DOI: 10.1242/dmm.050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
By controlling the passage of small molecules across lipid bilayers, membrane transporters influence not only the uptake and efflux of nutrients, but also the metabolic state of the cell. With more than 450 members, the Solute Carriers (SLCs) are the largest transporter super-family, clustering into families with different substrate specificities and regulatory properties. Cells of different types are, therefore, able to tailor their transporter expression signatures depending on their metabolic requirements, and the physiological importance of these proteins is illustrated by their mis-regulation in a number of disease states. In cancer, transporter expression is heterogeneous, and the SLC family has been shown to facilitate the accumulation of biomass, influence redox homeostasis, and also mediate metabolic crosstalk with other cell types within the tumour microenvironment. This Review explores the roles of membrane transporters in physiological and malignant settings, and how these roles can affect drug response, through either indirect modulation of sensitivity or the direct transport of small-molecule therapeutic compounds into cells.
Collapse
Affiliation(s)
- Sara Alam
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Emily Doherty
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paula Ortega-Prieto
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Julia Arizanova
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Louise Fets
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
10
|
Wang Z, Zhu H, Xiong W. Advances in mass spectrometry-based multi-scale metabolomic methodologies and their applications in biological and clinical investigations. Sci Bull (Beijing) 2023; 68:2268-2284. [PMID: 37666722 DOI: 10.1016/j.scib.2023.08.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Metabolomics is a nascent field of inquiry that emerged in the late 20th century. It encompasses the comprehensive profiling of metabolites across a spectrum of organisms, ranging from bacteria and cells to tissues. The rapid evolution of analytical methods and data analysis has greatly accelerated progress in this dynamic discipline over recent decades. Sophisticated techniques such as liquid chromatograph mass spectrometry (MS), gas chromatograph MS, capillary electrophoresis MS, and nuclear magnetic resonance serve as the cornerstone of metabolomic analysis. Building upon these methods, a plethora of modifications and combinations have emerged to propel the advancement of metabolomics. Despite this progress, scrutinizing metabolism at the single-cell or single-organelle level remains an arduous task over the decades. Some of the most thrilling advancements, such as single-cell and single-organelle metabolic profiling techniques, offer profound insights into the intricate mechanisms within cells and organelles. This allows for a comprehensive study of metabolic heterogeneity and its pivotal role in multiple biological processes. The progress made in MS imaging has enabled high-resolution in situ metabolic profiling of tissue sections and even individual cells. Spatial reconstruction techniques enable the direct representation of metabolic distribution and alteration in three-dimensional space. The application of novel metabolomic techniques has led to significant breakthroughs in biological and clinical studies, including the discovery of novel metabolic pathways, determination of cell fate in differentiation, anti-aging intervention through modulating metabolism, metabolomics-based clinicopathologic analysis, and surgical decision-making based on on-site intraoperative metabolic analysis. This review presents a comprehensive overview of both conventional and innovative metabolomic techniques, highlighting their applications in groundbreaking biological and clinical studies.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Hongying Zhu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; CAS Key Laboratory of Brain Function and Disease, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China.
| | - Wei Xiong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; CAS Key Laboratory of Brain Function and Disease, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China.
| |
Collapse
|
11
|
Shegay PV, Shatova OP, Zabolotneva AA, Shestopalov AV, Kaprin AD. Moonlight functions of glycolytic enzymes in cancer. Front Mol Biosci 2023; 10:1076138. [PMID: 37449059 PMCID: PMC10337784 DOI: 10.3389/fmolb.2023.1076138] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Since an extensive genome research has started, basic principle "one gene-one protein-one function" was significantly revised. Many proteins with more than one function were identified and characterized as "moonlighting" proteins, which activity depend not only on structural peculiarities but also on compartmentation and metabolic environment. It turned out that "housekeeping" glycolytic enzymes show important moonlight functions such as control of development, proliferation, apoptosis, migration, regulation of transcription and cell signaling. Glycolytic enzymes emerged very early in evolution and because of the limited content of genomes, they could be used as ancient regulators for intercellular and intracellular communication. The multifunctionality of the constitutively expressed enzymes began to serve cancer cell survival and growth. In the present review we discuss some moonlight functions of glycolytic enzymes that important for malignant transformation and tumor growth.
Collapse
Affiliation(s)
- Petr V. Shegay
- Federal State Budget Institution, National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Olga P. Shatova
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Biochemistry Department, Peoples’ Friendship University of Russia, Moscow, Russia
| | - Anastasia A. Zabolotneva
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- National Medical Research Centre for Endocrinology, Laboratory of Biochemistry of Signaling Pathways, Moscow, Russia
| | - Aleksandr V. Shestopalov
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- National Medical Research Centre for Endocrinology, Laboratory of Biochemistry of Signaling Pathways, Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrei D. Kaprin
- Federal State Budget Institution, National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Biochemistry Department, Peoples’ Friendship University of Russia, Moscow, Russia
| |
Collapse
|
12
|
McNeale D, Esquirol L, Okada S, Strampel S, Dashti N, Rehm B, Douglas T, Vickers C, Sainsbury F. Tunable In Vivo Colocalization of Enzymes within P22 Capsid-Based Nanoreactors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17705-17715. [PMID: 36995754 DOI: 10.1021/acsami.3c00971] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Virus-like particles (VLPs) derived from bacteriophage P22 have been explored as biomimetic catalytic compartments. In vivo colocalization of enzymes within P22 VLPs uses sequential fusion to the scaffold protein, resulting in equimolar concentrations of enzyme monomers. However, control over enzyme stoichiometry, which has been shown to influence pathway flux, is key to realizing the full potential of P22 VLPs as artificial metabolons. We present a tunable strategy for stoichiometric control over in vivo co-encapsulation of P22 cargo proteins, verified for fluorescent protein cargo by Förster resonance energy transfer. This was then applied to a two-enzyme reaction cascade. l-homoalanine, an unnatural amino acid and chiral precursor to several drugs, can be synthesized from the readily available l-threonine by the sequential activity of threonine dehydratase and glutamate dehydrogenase. We found that the loading density of both enzymes influences their activity, with higher activity found at lower loading density implying an impact of molecular crowding on enzyme activity. Conversely, increasing overall loading density by increasing the amount of threonine dehydratase can increase activity from the rate-limiting glutamate dehydrogenase. This work demonstrates the in vivo colocalization of multiple heterologous cargo proteins in a P22-based nanoreactor and shows that controlled stoichiometry of individual enzymes in an enzymatic cascade is required for the optimal design of nanoscale biocatalytic compartments.
Collapse
Affiliation(s)
- Donna McNeale
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
- CSIRO Future Science Platform in Synthetic Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Dutton Park, QLD 4102, Australia
| | - Lygie Esquirol
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
- CSIRO Land and Water, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Black Mountain, ACT 2601, Australia
| | - Shoko Okada
- CSIRO Land and Water, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Black Mountain, ACT 2601, Australia
| | - Shai Strampel
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
| | - Noor Dashti
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
| | - Bernd Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
| | - Trevor Douglas
- Department of Chemistry, Indiana University, Indiana University, Bloomington, Indiana 47405, United States
| | - Claudia Vickers
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
- CSIRO Future Science Platform in Synthetic Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Dutton Park, QLD 4102, Australia
- ARC Centre of Excellence in Synthetic Biology, Queensland University of Technology, Brisbane, QLD 4000, Australia
- School of Biological and Environmental Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Frank Sainsbury
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, QLD 4111, Australia
- CSIRO Future Science Platform in Synthetic Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Dutton Park, QLD 4102, Australia
| |
Collapse
|
13
|
Shatova OP, Shegay PV, Zabolotneva AA, Shestopalov AV, Kaprin AD. Evolutionary Acquisition of Multifunctionality by Glycolytic Enzymes. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s002209302301009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
14
|
Armstrong FA, Cheng B, Herold RA, Megarity CF, Siritanaratkul B. From Protein Film Electrochemistry to Nanoconfined Enzyme Cascades and the Electrochemical Leaf. Chem Rev 2022; 123:5421-5458. [PMID: 36573907 PMCID: PMC10176485 DOI: 10.1021/acs.chemrev.2c00397] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protein film electrochemistry (PFE) has given unrivalled insight into the properties of redox proteins and many electron-transferring enzymes, allowing investigations of otherwise ill-defined or intractable topics such as unstable Fe-S centers and the catalytic bias of enzymes. Many enzymes have been established to be reversible electrocatalysts when attached to an electrode, and further investigations have revealed how unusual dependences of catalytic rates on electrode potential have stark similarities with electronics. A special case, the reversible electrochemistry of a photosynthetic enzyme, ferredoxin-NADP+ reductase (FNR), loaded at very high concentrations in the 3D nanopores of a conducting metal oxide layer, is leading to a new technology that brings PFE to myriad enzymes of other classes, the activities of which become controlled by the primary electron exchange. This extension is possible because FNR-based recycling of NADP(H) can be coupled to a dehydrogenase, and thence to other enzymes linked in tandem by the tight channelling of cofactors and intermediates within the nanopores of the material. The earlier interpretations of catalytic wave-shapes and various analogies with electronics are thus extended to initiate a field perhaps aptly named "cascade-tronics", in which the flow of reactions along an enzyme cascade is monitored and controlled through an electrochemical analyzer. Unlike in photosynthesis where FNR transduces electron transfer and hydride transfer through the unidirectional recycling of NADPH, the "electrochemical leaf" (e-Leaf) can be used to drive reactions in both oxidizing and reducing directions. The e-Leaf offers a natural way to study how enzymes are affected by nanoconfinement and crowding, mimicking the physical conditions under which enzyme cascades operate in living cells. The reactions of the trapped enzymes, often at very high local concentration, are thus studied electrochemically, exploiting the potential domain to control rates and direction and the current-rate analogy to derive kinetic data. Localized NADP(H) recycling is very efficient, resulting in very high cofactor turnover numbers and new opportunities for controlling and exploiting biocatalysis.
Collapse
Affiliation(s)
- Fraser A. Armstrong
- Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Beichen Cheng
- Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Ryan A. Herold
- Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Clare F. Megarity
- Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Bhavin Siritanaratkul
- Stephenson Institute for Renewable Energy and the Department of Chemistry, University of Liverpool, Liverpool L69 7ZF, United Kingdom
| |
Collapse
|
15
|
Yin M, Lei QY. BCAT2-BCKDH metabolon maintains BCAA homeostasis. Nat Metab 2022; 4:1618-1619. [PMID: 36443521 DOI: 10.1038/s42255-022-00680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Miao Yin
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, School of Basic Medical Sciences, Key Laboratory of Breast Cancer in Shanghai, Shanghai Key Laboratory of Radiation Oncology, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Qun-Ying Lei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, School of Basic Medical Sciences, Key Laboratory of Breast Cancer in Shanghai, Shanghai Key Laboratory of Radiation Oncology, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Zhang S, Zhang R, Yan X, Fan K. Nanozyme-Based Artificial Organelles: An Emerging Direction for Artificial Organelles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202294. [PMID: 35869033 DOI: 10.1002/smll.202202294] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Artificial organelles are compartmentalized nanoreactors, in which enzymes or enzyme-mimic catalysts exhibit cascade catalytic activities to mimic the functions of natural organelles. Importantly, research on artificial organelles paves the way for the bottom-up design of synthetic cells. Due to the separation effect of microcompartments, the catalytic reactions of enzymes are performed without the influence of the surrounding medium. The current techniques for synthesizing artificial organelles rely on the strategies of encapsulating enzymes into vesicle-structured materials or reconstituting enzymes onto the microcompartment materials. However, there are still some problems including limited functions, unregulated activities, and difficulty in targeting delivery that hamper the applications of artificial organelles. The emergence of nanozymes (nanomaterials with enzyme-like activities) provides novel ideas for the fabrication of artificial organelles. Compared with natural enzymes, nanozymes are featured with multiple enzymatic activities, higher stability, easier to synthesize, lower cost, and excellent recyclability. Herein, the most recent advances in nanozyme-based artificial organelles are summarized. Moreover, the benefits of compartmental structures for the applications of nanozymes, as well as the functional requirements of microcompartment materials are also introduced. Finally, the potential applications of nanozyme-based artificial organelles in biomedicine and the related challenges are discussed.
Collapse
Affiliation(s)
- Shuai Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruofei Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
17
|
Borsley S, Leigh DA, Roberts BMW. Chemical fuels for molecular machinery. Nat Chem 2022; 14:728-738. [PMID: 35778564 DOI: 10.1038/s41557-022-00970-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Chemical reaction networks that transform out-of-equilibrium 'fuel' to 'waste' are the engines that power the biomolecular machinery of the cell. Inspired by such systems, autonomous artificial molecular machinery is being developed that functions by catalysing the decomposition of chemical fuels, exploiting kinetic asymmetry to harness energy released from the fuel-to-waste reaction to drive non-equilibrium structures and dynamics. Different aspects of chemical fuels profoundly influence their ability to power molecular machines. Here we consider the structure and properties of the fuels that biology has evolved and compare their features with those of the rudimentary synthetic chemical fuels that have so far been used to drive autonomous non-equilibrium molecular-level dynamics. We identify desirable, but context-specific, traits for chemical fuels together with challenges and opportunities for the design and invention of new chemical fuels to power synthetic molecular machinery and other dissipative nanoscale processes.
Collapse
Affiliation(s)
- Stefan Borsley
- Department of Chemistry, University of Manchester, Manchester, UK
| | - David A Leigh
- Department of Chemistry, University of Manchester, Manchester, UK.
| | | |
Collapse
|
18
|
Jain A, Zoncu R. Organelle transporters and inter-organelle communication as drivers of metabolic regulation and cellular homeostasis. Mol Metab 2022; 60:101481. [PMID: 35342037 PMCID: PMC9043965 DOI: 10.1016/j.molmet.2022.101481] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Spatial compartmentalization of metabolic pathways within membrane-separated organelles is key to the ability of eukaryotic cells to precisely regulate their biochemical functions. Membrane-bound organelles such as mitochondria, endoplasmic reticulum (ER) and lysosomes enable the concentration of metabolic precursors within optimized chemical environments, greatly accelerating the efficiency of both anabolic and catabolic reactions, enabling division of labor and optimal utilization of resources. However, metabolic compartmentalization also poses a challenge to cells because it creates spatial discontinuities that must be bridged for reaction cascades to be connected and completed. To do so, cells employ different methods to coordinate metabolic fluxes occurring in different organelles, such as membrane-localized transporters to facilitate regulated metabolite exchange between mitochondria and lysosomes, non-vesicular transport pathways via physical contact sites connecting the ER with both mitochondria and lysosomes, as well as localized regulatory signaling processes that coordinately regulate the activity of all these organelles. SCOPE OF REVIEW This review covers how cells use membrane transporters, membrane contact sites, and localized signaling pathways to mediate inter-organelle communication and coordinate metabolism. We also describe how disruption of inter-organelle communication is an emerging driver in a multitude of diseases, from cancer to neurodegeneration. MAJOR CONCLUSIONS Effective communication among organelles is essential to cellular health and function. Identifying the major molecular players involved in mediating metabolic coordination between organelles will further our understanding of cellular metabolism in health and lead us to design better therapeutics against dysregulated metabolism in disease.
Collapse
Affiliation(s)
- Aakriti Jain
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
19
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
20
|
Jolly BJ, Co NH, Davis AR, Diaconescu PL, Liu C. A generalized kinetic model for compartmentalization of organometallic catalysis. Chem Sci 2022; 13:1101-1110. [PMID: 35211276 PMCID: PMC8790775 DOI: 10.1039/d1sc04983f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/21/2021] [Indexed: 11/21/2022] Open
Abstract
Compartmentalization is an attractive approach to enhance catalytic activity by retaining reactive intermediates and mitigating deactivating pathways. Such a concept has been well explored in biochemical and more recently, organometallic catalysis to ensure high reaction turnovers with minimal side reactions. However, the scarcity of theoretical frameworks towards confined organometallic chemistry impedes broader utility for the implementation of compartmentalization. Herein, we report a general kinetic model and offer design guidance for a compartmentalized organometallic catalytic cycle. In comparison to a non-compartmentalized catalysis, compartmentalization is quantitatively shown to prevent the unwanted intermediate deactivation, boost the corresponding reaction efficiency (γ), and subsequently increase catalytic turnover frequency (TOF). The key parameter in the model is the volumetric diffusive conductance (FV) that describes catalysts' diffusion propensity across a compartment's boundary. Optimal values of FV for a specific organometallic chemistry are needed to achieve maximal values of γ and TOF. As illustrated in specific reaction examples, our model suggests that a tailored compartment design, including the use of nanomaterials, is needed to suit a specific organometallic catalytic cycle. This work provides justification and design principles for further exploration into compartmentalizing organometallics to enhance catalytic performance. The conclusions from this work are generally applicable to other catalytic systems that need proper design guidance in confinement and compartmentalization. Compartmentalization is an attractive approach to enhance catalytic activity by retaining reactive intermediates and mitigating deactivating pathways.![]()
Collapse
Affiliation(s)
- Brandon J. Jolly
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
| | - Nathalie H. Co
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
| | - Ashton R. Davis
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
| | - Paula L. Diaconescu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
| | - Chong Liu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Giaccagli MM, Gómez-Elías MD, Herzfeld JD, Marín-Briggiler CI, Cuasnicú PS, Cohen DJ, Da Ros VG. Capacitation-Induced Mitochondrial Activity Is Required for Sperm Fertilizing Ability in Mice by Modulating Hyperactivation. Front Cell Dev Biol 2021; 9:767161. [PMID: 34765607 PMCID: PMC8576324 DOI: 10.3389/fcell.2021.767161] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 01/22/2023] Open
Abstract
To become fully competent to fertilize an egg, mammalian sperm undergo a series of functional changes within the female tract, known as capacitation, that require an adequate supply and management of energy. However, the contribution of each ATP generating pathway to sustain the capacitation-associated changes remains unclear. Based on this, we investigated the role of mitochondrial activity in the acquisition of sperm fertilizing ability during capacitation in mice. For this purpose, the dynamics of the mitochondrial membrane potential (MMP) was studied by flow cytometry with the probe tetramethylrhodamine ethyl ester (TMRE). We observed a time-dependent increase in MMP only in capacitated sperm as well as a specific staining with the probe in the flagellar region where mitochondria are confined. The MMP rise was prevented when sperm were exposed to the mitochondrial uncoupler carbonyl cyanide m-chlorophenyl hydrazine (CCCP) or the protein kinase A (PKA) inhibitor H89 during capacitation, indicating that MMP increase is dependent on capacitation and H89-sensitive events. Results showed that whereas nearly all motile sperm were TMRE positive, immotile cells were mostly TMRE negative, supporting an association between high MMP and sperm motility. Furthermore, CCCP treatment during capacitation did not affect PKA substrate and tyrosine phosphorylations but produced a decrease in hyperactivation measured by computer assisted sperm analysis (CASA), similar to that observed after H89 exposure. In addition, CCCP inhibited the in vitro sperm fertilizing ability without affecting cumulus penetration and gamete fusion, indicating that the hyperactivation supported by mitochondrial function is needed mainly for zona pellucida penetration. Finally, complementary in vivo fertilization experiments further demonstrated the fundamental role of mitochondrial activity for sperm function. Altogether, our results show the physiological relevance of mitochondrial functionality for sperm fertilization competence.
Collapse
Affiliation(s)
- María Milagros Giaccagli
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Matías Daniel Gómez-Elías
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Jael Dafne Herzfeld
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Clara Isabel Marín-Briggiler
- Laboratorio de Biología Celular y Molecular de la Reproducción, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Patricia Sara Cuasnicú
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Débora Juana Cohen
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Vanina Gabriela Da Ros
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
22
|
Sustained enzymatic activity and flow in crowded protein droplets. Nat Commun 2021; 12:6293. [PMID: 34725341 PMCID: PMC8560906 DOI: 10.1038/s41467-021-26532-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/11/2021] [Indexed: 11/09/2022] Open
Abstract
Living cells harvest energy from their environments to drive the chemical processes that enable life. We introduce a minimal system that operates at similar protein concentrations, metabolic densities, and length scales as living cells. This approach takes advantage of the tendency of phase-separated protein droplets to strongly partition enzymes, while presenting minimal barriers to transport of small molecules across their interface. By dispersing these microreactors in a reservoir of substrate-loaded buffer, we achieve steady states at metabolic densities that match those of the hungriest microorganisms. We further demonstrate the formation of steady pH gradients, capable of driving microscopic flows. Our approach enables the investigation of the function of diverse enzymes in environments that mimic cytoplasm, and provides a flexible platform for studying the collective behavior of matter driven far from equilibrium.
Collapse
|
23
|
Stevens RP, Paudel SS, Johnson SC, Stevens T, Lee JY. Endothelial metabolism in pulmonary vascular homeostasis and acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2021; 321:L358-L376. [PMID: 34159794 PMCID: PMC8384476 DOI: 10.1152/ajplung.00131.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 12/27/2022] Open
Abstract
Capillary endothelial cells possess a specialized metabolism necessary to adapt to the unique alveolar-capillary environment. Here, we highlight how endothelial metabolism preserves the integrity of the pulmonary circulation by controlling vascular permeability, defending against oxidative stress, facilitating rapid migration and angiogenesis in response to injury, and regulating the epigenetic landscape of endothelial cells. Recent reports on single-cell RNA-sequencing reveal subpopulations of pulmonary capillary endothelial cells with distinctive reparative capacities, which potentially offer new insight into their metabolic signature. Lastly, we discuss broad implications of pulmonary vascular metabolism on acute respiratory distress syndrome, touching on emerging findings of endotheliitis in coronavirus disease 2019 (COVID-19) lungs.
Collapse
Affiliation(s)
- Reece P Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Sunita S Paudel
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Santina C Johnson
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama
- Department of Biomolecular Engineering, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Ji Young Lee
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, Alabama
- Division of Pulmonary and Critical Care Medicine, College of Medicine, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| |
Collapse
|
24
|
Alves LQ, Ruivo R, Valente R, Fonseca MM, Machado AM, Plön S, Monteiro N, García-Parraga D, Ruiz-Díaz S, Sánchez-Calabuig MJ, Gutiérrez-Adán A, Castro LFC. A drastic shift in the energetic landscape of toothed whale sperm cells. Curr Biol 2021; 31:3648-3655.e9. [PMID: 34171300 DOI: 10.1016/j.cub.2021.05.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/22/2022]
Abstract
Mammalian spermatozoa are a notable example of metabolic compartmentalization.1 Energy in the form of ATP production, vital for motility, capacitation, and fertilization, is subcellularly separated in sperm cells. While glycolysis provides a local, rapid, and low-yielding input of ATP along the flagellum fibrous sheath, oxidative phosphorylation (OXPHOS), far more efficient over a longer time frame, is concentrated in the midpiece mitochondria.2 The relative weight of glycolysis and OXPHOS pathways in sperm function is variable among species and sensitive to oxygen and substrate availability.3-5 Besides partitioning energy production, sperm cell energetics display an additional singularity: the occurrence of sperm-specific gene duplicates and alternative spliced variants, with conserved function but structurally bound to the flagellar fibrous sheath.6,7 The wider selective forces driving the compartmentalization and adaptability of this energy system in mammalian species remain largely unknown, much like the impact of ecosystem resource availability (e.g., carbohydrates, fatty acids, and proteins) and dietary adaptations in reproductive physiology traits.8 Here, we investigated the Cetacea, an iconic group of fully aquatic and carnivorous marine mammals, evolutionarily related to extant terrestrial herbivores.9 In this lineage, episodes of profound trait remodeling have been accompanied by clear genomic signatures.10-14 We show that toothed whales exhibit impaired sperm glycolysis, due to gene and exon erosion, and demonstrate that dolphin spermatozoa motility depends on endogenous fatty acid β-oxidation, but not carbohydrates. Such unique energetic rewiring substantiates the observation of large mitochondria in toothed whale spermatozoa and emphasizes the radical physiological reorganization imposed by the transition to a carbohydrate-depleted marine environment.
Collapse
Affiliation(s)
- Luís Q Alves
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - Raquel Ruivo
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - Raul Valente
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Miguel M Fonseca
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - André M Machado
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Stephanie Plön
- Department of Pathology, Stellenbosch University, PO Box 241, Cape Town 8000, South Africa
| | - Nuno Monteiro
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua do Campo Alegre, 4169-007 Porto, Portugal; CIBIO - Research Centre in Biodiversity and Genetic Resources, Campus Agrário de Vairão, Rua Padre Armando Quintas, 4485-661 Vairão, Portugal
| | - David García-Parraga
- Veterinary Services, L'Oceanográfic, Ciudad de las Artes y las Ciencias, Junta de Murs i Vals, s/n, 46013 Valencia, Spain
| | - Sara Ruiz-Díaz
- Departamento de Reproducción Animal, INIA, Av. Puerta de Hierro, 18, 28040 Madrid, Spain; Mistral Fertility Clinics S.L., Clínica Tambre, 28002 Madrid, Spain
| | - Maria J Sánchez-Calabuig
- Departamento de Reproducción Animal, INIA, Av. Puerta de Hierro, 18, 28040 Madrid, Spain; Department of Animal Medicine and Surgery, Faculty of Veterinary Science, University Complutense of Madrid, 28040 Madrid, Spain
| | - Alfonso Gutiérrez-Adán
- Departamento de Reproducción Animal, INIA, Av. Puerta de Hierro, 18, 28040 Madrid, Spain.
| | - L Filipe C Castro
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua do Campo Alegre, 4169-007 Porto, Portugal.
| |
Collapse
|
25
|
Liang R, Menon V, Qiu J, Arif T, Renuse S, Lin M, Nowak R, Hartmann B, Tzavaras N, Benson DL, Chipuk JE, Fribourg M, Pandey A, Fowler V, Ghaffari S. Mitochondrial localization and moderated activity are key to murine erythroid enucleation. Blood Adv 2021; 5:2490-2504. [PMID: 34032849 PMCID: PMC8152511 DOI: 10.1182/bloodadvances.2021004259] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/15/2021] [Indexed: 02/04/2023] Open
Abstract
Mammalian red blood cells (RBCs), which primarily contain hemoglobin, exemplify an elaborate maturation process, with the terminal steps of RBC generation involving extensive cellular remodeling. This encompasses alterations of cellular content through distinct stages of erythroblast maturation that result in the expulsion of the nucleus (enucleation) followed by the loss of mitochondria and all other organelles and a transition to anaerobic glycolysis. Whether there is any link between erythroid removal of the nucleus and the function of any other organelle, including mitochondria, remains unknown. Here we demonstrate that mitochondria are key to nuclear clearance. Using live and confocal microscopy and high-throughput single-cell imaging, we show that before nuclear polarization, mitochondria progressively move toward one side of maturing erythroblasts and aggregate near the nucleus as it extrudes from the cell, a prerequisite for enucleation to proceed. Although we found active mitochondrial respiration is required for nuclear expulsion, levels of mitochondrial activity identify distinct functional subpopulations, because terminally maturing erythroblasts with low relative to high mitochondrial membrane potential are at a later stage of maturation, contain greatly condensed nuclei with reduced open chromatin-associated acetylation histone marks, and exhibit higher enucleation rates. Lastly, to our surprise, we found that late-stage erythroblasts sustain mitochondrial metabolism and subsequent enucleation, primarily through pyruvate but independent of in situ glycolysis. These findings demonstrate the critical but unanticipated functions of mitochondria during the erythroblast enucleation process. They are also relevant to the in vitro production of RBCs as well as to disorders of the erythroid lineage.
Collapse
Affiliation(s)
- Raymond Liang
- Department of Cell, Developmental and Regenerative Biology
- Developmental and Stem Cell Biology Multidisciplinary Training, Graduate School of Biomedical Sciences
| | - Vijay Menon
- Department of Cell, Developmental and Regenerative Biology
| | - Jiajing Qiu
- Department of Cell, Developmental and Regenerative Biology
| | - Tasleem Arif
- Department of Cell, Developmental and Regenerative Biology
| | - Santosh Renuse
- Institute of Genetic Medicine, and
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Miao Lin
- Department of Cell, Developmental and Regenerative Biology
| | - Roberta Nowak
- Department of Cell and Molecular Biology, Scripps Research Institute, La Jolla, CA; and
| | | | | | | | - Jerry E Chipuk
- Department of Oncological Sciences
- Tisch Cancer Institute
| | | | | | - Velia Fowler
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Saghi Ghaffari
- Department of Cell, Developmental and Regenerative Biology
- Developmental and Stem Cell Biology Multidisciplinary Training, Graduate School of Biomedical Sciences
- Department of Oncological Sciences
- Tisch Cancer Institute
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
26
|
Jang S, Xuan Z, Lagoy RC, Jawerth LM, Gonzalez IJ, Singh M, Prashad S, Kim HS, Patel A, Albrecht DR, Hyman AA, Colón-Ramos DA. Phosphofructokinase relocalizes into subcellular compartments with liquid-like properties in vivo. Biophys J 2021; 120:1170-1186. [PMID: 32853565 PMCID: PMC8059094 DOI: 10.1016/j.bpj.2020.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 02/08/2023] Open
Abstract
Although much is known about the biochemical regulation of glycolytic enzymes, less is understood about how they are organized inside cells. We systematically examine the dynamic subcellular localization of glycolytic protein phosphofructokinase-1/PFK-1.1 in Caenorhabditis elegans. We determine that endogenous PFK-1.1 localizes to subcellular compartments in vivo. In neurons, PFK-1.1 forms phase-separated condensates near synapses in response to energy stress from transient hypoxia. Restoring animals to normoxic conditions results in cytosolic dispersion of PFK-1.1. PFK-1.1 condensates exhibit liquid-like properties, including spheroid shapes due to surface tension, fluidity due to deformations, and fast internal molecular rearrangements. Heterologous self-association domain cryptochrome 2 promotes formation of PFK-1.1 condensates and recruitment of aldolase/ALDO-1. PFK-1.1 condensates do not correspond to stress granules and might represent novel metabolic subcompartments. Our studies indicate that glycolytic protein PFK-1.1 can dynamically form condensates in vivo.
Collapse
Affiliation(s)
- SoRi Jang
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Zhao Xuan
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Ross C Lagoy
- Department of Biomedical Engineering and Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Louise M Jawerth
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ian J Gonzalez
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Milind Singh
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Shavanie Prashad
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Hee Soo Kim
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Avinash Patel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Dirk R Albrecht
- Department of Biomedical Engineering and Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Daniel A Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut; Instituto de Neurobiología, Universidad de Puerto Rico, San Juan, Puerto Rico.
| |
Collapse
|
27
|
Strazza PS, de Siqueira DVF, Leão RM. ATP-sensitive K + channels control the spontaneous firing of a glycinergic interneuron in the auditory brainstem. J Physiol 2021; 599:1611-1630. [PMID: 33369743 DOI: 10.1113/jp280233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/16/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Cartwheel neurons provide potent inhibition to fusiform neurons in the dorsal cochlear nucleus (DCN). Most cartwheel neurons fire action potentials spontaneously, but the ion channels responsible for this intrinsic activity are unknown. We investigated the ion channels responsible for the intrinsic firing of cartwheel neurons and the stable resting membrane potential found in a fraction of these neurons (quiet neurons). Among the ion channels controlling membrane potential of cartwheel neurons, the presence of open ATP-sensitive potassium channels (KATP ) is responsible for the existence of quiet neurons. Our results pinpoint KATP channel modulation as a critical factor controlling the firing of cartwheel neurons. Hence, it is a crucial channel influencing the balance of excitation and inhibition in the DCN. ABSTRACT Cartwheel neurons from the dorsal cochlear nucleus (DCN) are glycinergic interneurons and the primary source of inhibition on the fusiform neurons, the DCN's principal excitatory neuron. Most cartwheel neurons present spontaneous firing (active neurons), producing a steady inhibitory tone on fusiform neurons. In contrast, a small fraction of these neurons do not fire spontaneously (quiet neurons). Hyperactivity of fusiform neurons is seen in animals with behavioural evidence of tinnitus. Because of its relevance in controlling the excitability of fusiform neurons, we investigated the ion channels responsible for the spontaneous firing of cartwheel neurons in DCN slices from rats. We found that quiet neurons presented an outward conductance not seen in active neurons, which generates a stable resting potential. This current was sensitive to tolbutamide, an ATP-sensitive potassium channel (KATP ) antagonist. After inhibition with tolbutamide, quiet neurons start to fire spontaneously, while the active neurons were not affected. On the other hand, in active neurons, KATP agonist diazoxide activated a conductance similar to quiet neurons' KATP conductance and stopped spontaneous firing. According to the effect of KATP channels on cartwheel neuron firing, glycinergic neurotransmission in DCN was increased by tolbutamide and decreased by diazoxide. Our results reveal a role of KATP channels in controlling the spontaneous firing of neurons not involved in fuel homeostasis.
Collapse
Affiliation(s)
- Paulo S Strazza
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Daniela V F de Siqueira
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo M Leão
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
28
|
Wentz JM, Bortz DM. BOUNDEDNESS OF A CLASS OF SPATIALLY DISCRETE REACTION-DIFFUSION SYSTEMS. SIAM JOURNAL ON APPLIED MATHEMATICS 2021; 81:1870-1892. [PMID: 38223745 PMCID: PMC10786630 DOI: 10.1137/20m131850x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Although the spatially discrete reaction-diffusion equation is often used to describe biological processes, the effect of diffusion in this framework is not fully understood. In the spatially continuous case, the incorporation of diffusion can cause blow-up with respect to the L ∞ norm, and criteria exist to determine whether the system is bounded for all time. However, no equivalent criteria exist for the discrete reaction-diffusion system. Due to the possible dynamical differences between these two system types and the advantage of using the spatially discrete representation to describe biological processes, it is worth examining the discrete system independently of the continuous system. Therefore, the focus of this paper is on determining sufficient conditions to guarantee that the discrete reaction-diffusion system is bounded for all time. We consider reaction-diffusion systems on a 1D domain with homogeneous Neumann boundary conditions and nonnegative initial data and solutions. We define a Lyapunov-like function and show that its existence guarantees that the discrete reaction-diffusion system is bounded. These results are considered in the context of four example systems for which Lyapunov-like functions can and cannot be found.
Collapse
Affiliation(s)
- Jacqueline M Wentz
- Department of Applied Mathematics, University of Colorado Boulder, Boulder, CO 80309 USA
| | - David M Bortz
- Department of Applied Mathematics, University of Colorado Boulder, Boulder, CO 80309 USA
| |
Collapse
|
29
|
Boon R, Silveira GG, Mostoslavsky R. Nuclear metabolism and the regulation of the epigenome. Nat Metab 2020; 2:1190-1203. [PMID: 33046909 DOI: 10.1038/s42255-020-00285-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022]
Abstract
Cellular metabolism has emerged as a major biological node governing cellular behaviour. Metabolic pathways fuel cellular energy needs, providing basic chemical molecules to sustain cellular homeostasis, proliferation and function. Changes in nutrient consumption or availability therefore can result in complete reprogramming of cellular metabolism towards stabilizing core metabolite pools, such as ATP, S-adenosyl methionine, acetyl-CoA, NAD/NADP and α-ketoglutarate. Because these metabolites underlie a variety of essential metabolic reactions, metabolism has evolved to operate in separate subcellular compartments through diversification of metabolic enzyme complexes, oscillating metabolic activity and physical separation of metabolite pools. Given that these same core metabolites are also consumed by chromatin modifiers in the establishment of epigenetic signatures, metabolite consumption on and release from chromatin directly influence cellular metabolism and gene expression. In this Review, we highlight recent studies describing the mechanisms determining nuclear metabolism and governing the redistribution of metabolites between the nuclear and non-nuclear compartments.
Collapse
Affiliation(s)
- Ruben Boon
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Giorgia G Silveira
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Raul Mostoslavsky
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
30
|
S Mogre S, Brown AI, Koslover EF. Getting around the cell: physical transport in the intracellular world. Phys Biol 2020; 17:061003. [PMID: 32663814 DOI: 10.1088/1478-3975/aba5e5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Eukaryotic cells face the challenging task of transporting a variety of particles through the complex intracellular milieu in order to deliver, distribute, and mix the many components that support cell function. In this review, we explore the biological objectives and physical mechanisms of intracellular transport. Our focus is on cytoplasmic and intra-organelle transport at the whole-cell scale. We outline several key biological functions that depend on physically transporting components across the cell, including the delivery of secreted proteins, support of cell growth and repair, propagation of intracellular signals, establishment of organelle contacts, and spatial organization of metabolic gradients. We then review the three primary physical modes of transport in eukaryotic cells: diffusive motion, motor-driven transport, and advection by cytoplasmic flow. For each mechanism, we identify the main factors that determine speed and directionality. We also highlight the efficiency of each transport mode in fulfilling various key objectives of transport, such as particle mixing, directed delivery, and rapid target search. Taken together, the interplay of diffusion, molecular motors, and flows supports the intracellular transport needs that underlie a broad variety of biological phenomena.
Collapse
Affiliation(s)
- Saurabh S Mogre
- Department of Physics, University of California, San Diego, San Diego, California 92093, United States of America
| | | | | |
Collapse
|
31
|
Belluati A, Craciun I, Palivan CG. Bioactive Catalytic Nanocompartments Integrated into Cell Physiology and Their Amplification of a Native Signaling Cascade. ACS NANO 2020; 14:12101-12112. [PMID: 32869973 DOI: 10.1021/acsnano.0c05574] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Bioactive nanomaterials have the potential to overcome the limitations of classical pharmacological approaches by taking advantage of native pathways to influence cell behavior, interacting with them and eliciting responses. Herein, we propose a cascade system mediated by two catalytic nanocompartments (CNC) with biological activity. Activated by nitric oxide (NO) produced by inducible nitric oxidase synthase (iNOS), soluble guanylyl cyclase (sGC) produces cyclic guanosine monophosphate (cGMP), a second messenger that modulates a broad range of physiological functions. As alterations in cGMP signaling are implicated in a multitude of pathologies, its signaling cascade represents a viable target for therapeutic intervention. Following along this line, we encapsulated iNOS and sGC in two separate polymeric compartments that function in unison to produce NO and cGMP. Their action was tested in vitro by monitoring the derived changes in cytoplasmic calcium concentrations of HeLa and differentiated C2C12 myocytes, where the produced second messenger influenced the cellular homeostasis.
Collapse
Affiliation(s)
- Andrea Belluati
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058 Basel, Switzerland
| |
Collapse
|
32
|
Huttanus HM, Senger RS. A synthetic biosensor to detect peroxisomal acetyl-CoA concentration for compartmentalized metabolic engineering. PeerJ 2020; 8:e9805. [PMID: 33194349 PMCID: PMC7485502 DOI: 10.7717/peerj.9805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/03/2020] [Indexed: 11/20/2022] Open
Abstract
Background Sub-cellular compartmentalization is used by cells to create favorable microenvironments for various metabolic reactions. These compartments concentrate enzymes, separate competing metabolic reactions, and isolate toxic intermediates. Such advantages have been recently harnessed by metabolic engineers to improve the production of various high-value chemicals via compartmentalized metabolic engineering. However, measuring sub-cellular concentrations of key metabolites represents a grand challenge for compartmentalized metabolic engineering. Methods To this end, we developed a synthetic biosensor to measure a key metabolite, acetyl-CoA, in a representative compartment of yeast, the peroxisome. This synthetic biosensor uses enzyme re-localization via PTS1 signal peptides to construct a metabolic pathway in the peroxisome which converts acetyl-CoA to polyhydroxybutyrate (PHB) via three enzymes. The PHB is then quantified by HPLC. Results The biosensor demonstrated the difference in relative peroxisomal acetyl-CoA availability under various culture conditions and was also applied to screening a library of single knockout yeast mutants. The screening identified several mutants with drastically reduced peroxisomal acetyl-CoA and one with potentially increased levels. We expect our synthetic biosensors can be widely used to investigate sub-cellular metabolism and facilitate the “design-build-test” cycle of compartmentalized metabolic engineering.
Collapse
Affiliation(s)
- Herbert M Huttanus
- Department of Biological Systems Engineering, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, VA, United States of America
| | - Ryan S Senger
- Department of Biological Systems Engineering, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, VA, United States of America.,Department of Chemical Engineering, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, VA, United States of America
| |
Collapse
|
33
|
Diehl KL, Muir TW. Chromatin as a key consumer in the metabolite economy. Nat Chem Biol 2020; 16:620-629. [PMID: 32444835 DOI: 10.1038/s41589-020-0517-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/05/2020] [Indexed: 12/31/2022]
Abstract
In eukaryotes, chromatin remodeling and post-translational modifications (PTMs) shape the local chromatin landscape to establish permissive and repressive regions within the genome, orchestrating transcription, replication, and DNA repair in concert with other epigenetic mechanisms. Though cellular nutrient signaling encompasses a huge number of pathways, recent attention has turned to the hypothesis that the metabolic state of the cell is communicated to the genome through the type and concentration of metabolites in the nucleus that are cofactors for chromatin-modifying enzymes. Importantly, both epigenetic and metabolic dysregulation are hallmarks of a range of diseases, and this metabolism-chromatin axis may yield a well of new therapeutic targets. In this Perspective, we highlight emerging themes in the inter-regulation of the genome and metabolism via chromatin, including nonenzymatic histone modifications arising from chemically reactive metabolites, the expansion of PTM diversity from cofactor-promiscuous chromatin-modifying enzymes, and evidence for the existence and importance of subnucleocytoplasmic metabolite pools.
Collapse
Affiliation(s)
- Katharine L Diehl
- Department of Chemistry, Princeton University, Princeton, NJ, USA. .,Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT, USA.
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
34
|
Krishnan J, Lu L, Alam Nazki A. The interplay of spatial organization and biochemistry in building blocks of cellular signalling pathways. J R Soc Interface 2020; 17:20200251. [PMID: 32453980 PMCID: PMC7276544 DOI: 10.1098/rsif.2020.0251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022] Open
Abstract
Biochemical pathways and networks are central to cellular information processing. While a broad range of studies have dissected multiple aspects of information processing in biochemical pathways, the effect of spatial organization remains much less understood. It is clear that space is central to intracellular organization, plays important roles in cellular information processing and has been exploited in evolution; additionally, it is being increasingly exploited in synthetic biology through the development of artificial compartments, in a variety of ways. In this paper, we dissect different aspects of the interplay between spatial organization and biochemical pathways, by focusing on basic building blocks of these pathways: covalent modification cycles and two-component systems, with enzymes which may be monofunctional or bifunctional. Our analysis of spatial organization is performed by examining a range of 'spatial designs': patterns of localization or non-localization of enzymes/substrates, theoretically and computationally. Using these well-characterized in silico systems, we analyse the following. (i) The effect of different types of spatial organization on the overall kinetics of modification, and the role of distinct modification mechanisms therein. (ii) How different information processing characteristics seen experimentally and studied from the viewpoint of kinetics are perturbed, or generated. (iii) How the activity of enzymes (bifunctional enzymes in particular) may be spatially manipulated, and the relationship between localization and activity. (iv) How transitions in spatial organization (encountered either through evolution or through the lifetime of cells, as seen in multiple model organisms) impacts the kinetic module (and pathway) behaviour, and how transitions in chemistry may be impacted by prior spatial organization. The basic insights which emerge are central to understanding the role of spatial organization in biochemical pathways in both bacteria and eukaryotes, and are of direct relevance to engineering spatial organization of pathways in bottom-up synthetic biology in cellular and cell-free systems.
Collapse
Affiliation(s)
- J. Krishnan
- Department of Chemical Engineering, Centre for Process Systems Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
- Institute for Systems and Synthetic Biology, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Lingjun Lu
- Department of Chemical Engineering, Centre for Process Systems Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Aiman Alam Nazki
- Department of Chemical Engineering, Centre for Process Systems Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|
35
|
Abstract
Skeletal involvement is a frequent and troublesome complication in advanced cancers. In the process of tumor cells homing to the skeleton to form bone metastases (BM), different mechanisms allow tumor cells to interact with cells of the bone microenvironment and seed in the bone tissue. Among these, tumor acidosis has been directly associated with tumor invasion and aggressiveness in several types of cancer although it has been less explored in the context of BM. In bone, the association of local acidosis and cancer invasiveness is even more important for tumor expansion since the extracellular matrix is formed by both organic and hard inorganic matrices and bone cells are used to sense protons and adapt or react to a low pH to maintain tissue homeostasis. In the BM microenvironment, increased concentration of protons may derive not only from glycolytic tumor cells but also from tumor-induced osteoclasts, the bone-resorbing cells, and may influence the progression or symptoms of BM in many different ways, by directly enhancing cancer cell motility and aggressiveness, or by modulating the functions of bone cells versus a pro-tumorigenic phenotype, or by inducing bone pain. In this review, we will describe and discuss the cause of acidosis in BM, its role in BM microenvironment, and which are the final effectors that may be targeted to treat metastatic patients.
Collapse
Affiliation(s)
- Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Gemma Di Pompo
- Orthopaedic Pathophysiology and Regenerative Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Silvia Lemma
- Orthopaedic Pathophysiology and Regenerative Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40123, Bologna, Italy
| |
Collapse
|
36
|
Fernández-García J, Altea-Manzano P, Pranzini E, Fendt SM. Stable Isotopes for Tracing Mammalian-Cell Metabolism In Vivo. Trends Biochem Sci 2020; 45:185-201. [PMID: 31955965 DOI: 10.1016/j.tibs.2019.12.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023]
Abstract
Metabolism is at the cornerstone of all cellular functions and mounting evidence of its deregulation in different diseases emphasizes the importance of a comprehensive understanding of metabolic regulation at the whole-organism level. Stable-isotope measurements are a powerful tool for probing cellular metabolism and, as a result, are increasingly used to study metabolism in in vivo settings. The additional complexity of in vivo metabolic measurements requires paying special attention to experimental design and data interpretation. Here, we review recent work where in vivo stable-isotope measurements have been used to address relevant biological questions within an in vivo context, summarize different experimental and data interpretation approaches and their limitations, and discuss future opportunities in the field.
Collapse
Affiliation(s)
- Juan Fernández-García
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium.
| | - Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Erica Pranzini
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium; Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
37
|
Germeys C, Vandoorne T, Bercier V, Van Den Bosch L. Existing and Emerging Metabolomic Tools for ALS Research. Genes (Basel) 2019; 10:E1011. [PMID: 31817338 PMCID: PMC6947647 DOI: 10.3390/genes10121011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/23/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that aberrant energy metabolism could play an important role in the pathogenesis of amyotrophic lateral sclerosis (ALS). Despite this, studies applying advanced technologies to investigate energy metabolism in ALS remain scarce. The rapidly growing field of metabolomics offers exciting new possibilities for ALS research. Here, we review existing and emerging metabolomic tools that could be used to further investigate the role of metabolism in ALS. A better understanding of the metabolic state of motor neurons and their surrounding cells could hopefully result in novel therapeutic strategies.
Collapse
Affiliation(s)
- Christine Germeys
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Tijs Vandoorne
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Valérie Bercier
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| |
Collapse
|
38
|
Belluati A, Craciun I, Meyer CE, Rigo S, Palivan CG. Enzymatic reactions in polymeric compartments: nanotechnology meets nature. Curr Opin Biotechnol 2019; 60:53-62. [DOI: 10.1016/j.copbio.2018.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/14/2018] [Accepted: 12/19/2018] [Indexed: 01/28/2023]
|
39
|
Wellen KE, Snyder NW. Should we consider subcellular compartmentalization of metabolites, and if so, how do we measure them? Curr Opin Clin Nutr Metab Care 2019; 22:347-354. [PMID: 31365463 PMCID: PMC6824478 DOI: 10.1097/mco.0000000000000580] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW To examine the consequences of metabolism compartmentalized at the subcellular level, provide prototypical examples of compartmentalized metabolism, and describe methods to examine compartmentalized metabolism. RECENT FINDINGS Progress in metabolomics and isotope tracing has underscored the importance of subcellular compartments of metabolism. The discovery of biological effects of metabolites as bioenergetic intermediates, anabolic building blocks, signaling mediators, and effectors in posttranslation modifications of proteins and nucleic acids have highlighted the role of compartmentalization in determining metabolic fate. Recent advances in both direct and indirect methods to quantify compartmentalized metabolism have improved upon historical approaches. Genetically encoded metabolite sensors, chemical probes, immunoaffinity purification, and compartment-resolved metabolic modeling have all been recently applied to study compartmentalization. SUMMARY Accurate measurement of metabolites in distinct subcellular compartments is important for understanding and pharmacologically targeting metabolic pathways in diverse disease contexts, including cancer, diabetes, heart failure, obesity, and regulation of the immune system. Direct and indirect approaches to quantify compartmentalized metabolism are advancing rapidly. Yet, major challenges remain in the generalizability, rigor, and interpretation of data from the available methods to quantify compartmentalized metabolism.
Collapse
Affiliation(s)
- Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
40
|
Svoboda P, Krizova E, Sestakova S, Vapenkova K, Knejzlik Z, Rimpelova S, Rayova D, Volfova N, Krizova I, Rumlova M, Sykora D, Kizek R, Haluzik M, Zidek V, Zidkova J, Skop V. Nuclear transport of nicotinamide phosphoribosyltransferase is cell cycle-dependent in mammalian cells, and its inhibition slows cell growth. J Biol Chem 2019; 294:8676-8689. [PMID: 30975903 DOI: 10.1074/jbc.ra118.003505] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 04/04/2019] [Indexed: 01/26/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is located in both the nucleus and cytoplasm and has multiple biological functions including catalyzing the rate-limiting step in NAD synthesis. Moreover, up-regulated NAMPT expression has been observed in many cancers. However, the determinants and regulation of NAMPT's nuclear transport are not known. Here, we constructed a GFP-NAMPT fusion protein to study NAMPT's subcellular trafficking. We observed that in unsynchronized 3T3-L1 preadipocytes, 25% of cells had higher GFP-NAMPT fluorescence in the cytoplasm, and 62% had higher GFP-NAMPT fluorescence in the nucleus. In HepG2 hepatocytes, 6% of cells had higher GFP-NAMPT fluorescence in the cytoplasm, and 84% had higher GFP-NAMPT fluorescence in the nucleus. In both 3T3-L1 and HepG2 cells, GFP-NAMPT was excluded from the nucleus immediately after mitosis and migrated back into it as the cell cycle progressed. In HepG2 cells, endogenous, untagged NAMPT displayed similar changes with the cell cycle, and in nonmitotic cells, GFP-NAMPT accumulated in the nucleus. Similarly, genotoxic, oxidative, or dicarbonyl stress also caused nuclear NAMPT localization. These interventions also increased poly(ADP-ribosyl) polymerase and sirtuin activity, suggesting an increased cellular demand for NAD. We identified a nuclear localization signal in NAMPT and amino acid substitution in this sequence (424RSKK to ASGA), which did not affect its enzymatic activity, blocked nuclear NAMPT transport, slowed cell growth, and increased histone H3 acetylation. These results suggest that NAMPT is transported into the nucleus where it presumably increases NAD synthesis required for cell proliferation. We conclude that specific inhibition of NAMPT transport into the nucleus might be a potential avenue for managing cancer.
Collapse
Affiliation(s)
- Petr Svoboda
- From the Departments of Biochemistry and Microbiology.,the Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Czech Republic
| | - Edita Krizova
- From the Departments of Biochemistry and Microbiology
| | | | | | | | | | - Diana Rayova
- From the Departments of Biochemistry and Microbiology
| | - Nikol Volfova
- From the Departments of Biochemistry and Microbiology
| | | | | | - David Sykora
- Analytical Chemistry, University of Chemistry and Technology Prague, Prague 6, 166 28, Czech Republic
| | - Rene Kizek
- the Department of Human Pharmacology and Toxicology, University of Veterinary and Pharmaceutical Sciences Brno, Brno, 612 42, Czech Republic
| | - Martin Haluzik
- the Centre for Experimental Medicine and.,Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague 4, 140 21, Czech Republic, and.,the Institute of Medical Biochemistry and Laboratory Diagnostics, Charles University and General University Hospital in Prague, Prague 2, 128 08, Czech Republic
| | - Vaclav Zidek
- the Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Czech Republic
| | | | - Vojtech Skop
- From the Departments of Biochemistry and Microbiology, .,the Centre for Experimental Medicine and
| |
Collapse
|
41
|
De Bernardis Murat C, Leão RM. A voltage-dependent depolarization induced by low external glucose in neurons of the nucleus of the tractus solitarius: interaction with K ATP channels. J Physiol 2019; 597:2515-2532. [PMID: 30927460 DOI: 10.1113/jp277729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/21/2019] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Neurons from the brainstem nucleus of the tractus solitarius (NTS) participate in the counter-regulatory mechanisms in response to hypoglycaemia. ATP-sensitive potassium (KATP ) channels are expressed in NTS neurons, and are partially open at rest in normoglycaemic 5 mM glucose. In normoglycaemic conditions, most NTS neurons depolarize in response to low external glucose (0.5 mM), via a voltage-dependent mechanism. Conversely, most NTS neurons incubated in hyperglycaemic 10 mM glucose do not respond to low glucose due to a more positive resting membrane potential caused by the closure of KATP channels following increased intracellular metabolic ATP. Our findings show that in hyperglycaemic conditions, NTS neurons failed to sense rapid changes in external glucose, which could be related to hypoglycaemia-associated autonomic failure. ABSTRACT The nucleus of the tractus solitarius (NTS) is an integrative centre for autonomic counter-regulatory responses to hypoglycaemia. KATP channels link the metabolic status of the neuron to its excitability. Here we investigated the influence of KATP channels on the membrane potential of NTS neurons in normo- and hyperglycaemic external glucose concentrations, and after switching to a hypoglycaemic concentration, using in vitro electrophysiological recordings in brainstem slices. We found that in normoglycaemic (5 mM) glucose, tolbutamide, a KATP channel antagonist, depolarized the membrane of most neurons, and this effect was observed in more hyperpolarized neurons. All neurons hyperpolarized after pharmacological activation of KATP channels. Most NTS neurons depolarized in the presence of low glucose (0.5 mM), and this effect was only seen in hyperpolarized neurons. The effect of glucose was caused by a cationic current with a reversal potential around -50 mV. In the presence of hyperglycaemic glucose (10 mM), neurons were more depolarized, and fewer neurons responded to KATP blockage. Application of 0.5 mM glucose solution to these neurons depolarized the membrane only in more hyperpolarized neurons. We conclude that NTS neurons present with KATP channels open at rest in normoglycaemic conditions, and their membrane potential is affected by extracellular glucose. Moreover, NTS neurons depolarize the membrane in response to the application of a low glucose solution, but this effect is occluded by membrane depolarization triggered by KATP blockage. Our data suggest a homeostatic regulation of the membrane potential by external glucose, and a possible mechanism related to the hypoglycaemia-associated autonomic failure.
Collapse
Affiliation(s)
- Cahuê De Bernardis Murat
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo Mauricio Leão
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
42
|
Zerfaß C, Asally M, Soyer OS. Interrogating metabolism as an electron flow system. CURRENT OPINION IN SYSTEMS BIOLOGY 2019; 13:59-67. [PMID: 31008413 PMCID: PMC6472609 DOI: 10.1016/j.coisb.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolism is generally considered as a neatly organised system of modular pathways, shaped by evolution under selection for optimal cellular growth. This view falls short of explaining and predicting a number of key observations about the structure and dynamics of metabolism. We highlight these limitations of a pathway-centric view on metabolism and summarise studies suggesting how these could be overcome by viewing metabolism as a thermodynamically and kinetically constrained, dynamical flow system. Such a systems-level, first-principles based view of metabolism can open up new avenues of metabolic engineering and cures for metabolic diseases and allow better insights to a myriad of physiological processes that are ultimately linked to metabolism. Towards further developing this view, we call for a closer interaction among physical and biological disciplines and an increased use of electrochemical and biophysical approaches to interrogate cellular metabolism together with the microenvironment in which it exists.
Collapse
Affiliation(s)
- Christian Zerfaß
- Bio-Electrical Engineering (BEE) Innovation Hub, University of Warwick, Coventry, CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Munehiro Asally
- Bio-Electrical Engineering (BEE) Innovation Hub, University of Warwick, Coventry, CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, CV4 7AL, UK
| | - Orkun S. Soyer
- Bio-Electrical Engineering (BEE) Innovation Hub, University of Warwick, Coventry, CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
43
|
Affiliation(s)
- Stanislav Tsitkov
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Henry Hess
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
44
|
Uhrich D, Jang HY, Park JB, von Langermann J. Characterization and application of chemical-resistant polyurethane-based enzyme and whole cell compartments. J Biotechnol 2019; 289:31-38. [PMID: 30439386 DOI: 10.1016/j.jbiotec.2018.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/23/2018] [Accepted: 11/07/2018] [Indexed: 01/28/2023]
Abstract
This study presents the preparation and physical-chemical characterization of chemical resistant polyurethane-based compartments for biocatalytic application. The artificial compartments were prepared from an emulsion of polymer precursor and an aqueous phase that includes a biocatalytic reaction system. After curing, highly dispersed aqueous domains were obtained, which still contain the entire biocatalytic reaction system and remain fixed in the solid polymer preparation. The tensile and compression behavior of the prepared polymeric material is not significantly affected by the incorporation and facilitates excellent stability against various organic solvents and acid solutions. Thereby, the compartments can be used not only for enantioselective alcohol-dehydrogenase catalyzed reduction but also for a whole cell catalyzed hydrolysis of esters. Moreover, the compartmented whole-cell system was considerably stable to allow multiple reuses without a noticeable loss of catalytic activity of the incorporated whole cell catalytic reaction system.
Collapse
Affiliation(s)
- Diana Uhrich
- Biocatalytic Synthesis Group, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Hyun-Young Jang
- Department of Food Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - Jin-Byung Park
- Department of Food Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - Jan von Langermann
- Biocatalytic Synthesis Group, Institute of Chemistry, University of Rostock, Rostock, Germany.
| |
Collapse
|
45
|
Agrawal A, Pekkurnaz G, Koslover EF. Spatial control of neuronal metabolism through glucose-mediated mitochondrial transport regulation. eLife 2018; 7:40986. [PMID: 30561333 PMCID: PMC6322862 DOI: 10.7554/elife.40986] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023] Open
Abstract
Eukaryotic cells modulate their metabolism by organizing metabolic components in response to varying nutrient availability and energy demands. In rat axons, mitochondria respond to glucose levels by halting active transport in high glucose regions. We employ quantitative modeling to explore physical limits on spatial organization of mitochondria and localized metabolic enhancement through regulated stopping of processive motion. We delineate the role of key parameters, including cellular glucose uptake and consumption rates, that are expected to modulate mitochondrial distribution and metabolic response in spatially varying glucose conditions. Our estimates indicate that physiological brain glucose levels fall within the limited range necessary for metabolic enhancement. Hence mitochondrial localization is shown to be a plausible regulatory mechanism for neuronal metabolic flexibility in the presence of spatially heterogeneous glucose, as may occur in long processes of projection neurons. These findings provide a framework for the control of cellular bioenergetics through organelle trafficking. Cells are equipped with power factories called mitochondria that turn nutrients into chemical energy to fuel processes in the cell. Hundreds of mitochondria move throughout the cell, shifting their positions in response to energy demands. This happens via molecular motors that pick the mitochondria up and carry them to new locations. Such movements enable the mitochondria to accumulate in parts of the cell with the greatest energy needs. Mitochondria of nerve cells or neurons have a particular challenging job, as neurons can be very long and different parts within the cells can have different energy needs. It has been shown that mitochondria stop in regions where nutrients such as sugar are most concentrated. So far, it has been unclear whether this regulated stopping helps control energy balance in neurons. Here, Agrawal et al. used a computational model of rat neurons to find out whether sugar levels are sufficient in guiding mitochondria. The results showed that the mitochondria only accumulated in high-nutrient regions when the sugar concentrations were moderate – not too low and not too high. A specific range of sugar levels was necessary to make this mechanism useful for increasing the efficiency of energy production. Such concentrations match the ones observed in healthy rat brains. When neurons are unable to meet their energy demands, they stop working and sometimes even die. This is the case in many diseases, including diabetes, dementia, and Alzheimer’s disease. Computer models allow us to explore the complex energy regulation in detail. A better understanding of how neurons regulate their energy production and demand may help us discover how they become faulty in these diseases.
Collapse
Affiliation(s)
- Anamika Agrawal
- Department of Physics, University of California, San Diego, San Diego, United States
| | - Gulcin Pekkurnaz
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - Elena F Koslover
- Department of Physics, University of California, San Diego, San Diego, United States
| |
Collapse
|
46
|
Niu X, Chen YJ, Crawford PA, Patti GJ. Transport-exclusion pharmacology to localize lactate dehydrogenase activity within cells. Cancer Metab 2018; 6:19. [PMID: 30559963 PMCID: PMC6290536 DOI: 10.1186/s40170-018-0192-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/24/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Recent in vitro and in vivo work has shown that lactate provides an important source of carbon for metabolic reactions in cancer cell mitochondria. An interesting question is whether lactate is oxidized by lactate dehydrogenase (LDH) in the cytosol and/or in mitochondria. Since metabolic processes in the cytosol and mitochondria are affected by redox balance, the location of LDH may have important regulatory implications in cancer metabolism. METHODS Within most mammalian cells, metabolic processes are physically separated by membrane-bound compartments. Our general understanding of this spatial organization and its role in cellular function, however, suffers from the limited number of techniques to localize enzymatic activities within a cell. Here, we describe an approach to assess metabolic compartmentalization by monitoring the activity of pharmacological inhibitors that cannot be transported into specific cellular compartments. RESULTS Oxamate, which chemically resembles pyruvate, is transported into mitochondria and inhibits LDH activity in purified mitochondria. GSK-2837808A, in contrast, is a competitive inhibitor of NAD, which cannot cross the inner mitochondrial membrane. GSK-2837808A did not inhibit the LDH activity of intact mitochondria, but GSK-2837808A did inhibit LDH activity after the inner mitochondrial membrane was disrupted. CONCLUSIONS Our results are consistent with some mitochondrial LDH that is accessible to oxamate, but inaccessible to GSK-2837808A until mitochondria are homogenized. This strategy of using inhibitors with selective access to subcellular compartments, which we refer to as transport-exclusion pharmacology, is broadly applicable to localize other metabolic reactions within cells.
Collapse
Affiliation(s)
- Xiangfeng Niu
- Department of Chemistry, Washington University, St. Louis, USA
| | - Ying-Jr Chen
- Department of Chemistry, Washington University, St. Louis, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Gary J. Patti
- Department of Chemistry, Washington University, St. Louis, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, USA
| |
Collapse
|
47
|
Enzymatic complexes across scales. Essays Biochem 2018; 62:501-514. [PMID: 30315098 PMCID: PMC6204551 DOI: 10.1042/ebc20180008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023]
Abstract
An unprecedented opportunity to integrate ~100 years of meticulous in vitro biomolecular research is currently provided in the light of recent advances in methods to visualize closer-to-native architectures of biomolecular machines, and metabolic enzymes in particular. Traditional views of enzymes, namely biomolecular machines, only partially explain their role, organization and kinetics in the cellular milieu. Enzymes self- or hetero-associate, form fibers, may bind to membranes or cytoskeletal elements, have regulatory roles, associate into higher order assemblies (metabolons) or even actively participate in phase-separated membraneless organelles, and all the above in a transient, temporal and spatial manner in response to environmental changes or structural/functional changes of their assemblies. Here, we focus on traditional and emerging concepts in cellular biochemistry and discuss new opportunities in bridging structural, molecular and cellular analyses for metabolic pathways, accumulated over the years, highlighting functional aspects of enzymatic complexes discussed across different levels of spatial resolution.
Collapse
|
48
|
Glycolytic metabolism is essential for CCR7 oligomerization and dendritic cell migration. Nat Commun 2018; 9:2463. [PMID: 29941886 PMCID: PMC6018630 DOI: 10.1038/s41467-018-04804-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 05/15/2018] [Indexed: 02/04/2023] Open
Abstract
Dendritic cells (DCs) are first responders of the innate immune system that integrate signals from external stimuli to direct context-specific immune responses. Current models suggest that an active switch from mitochondrial metabolism to glycolysis accompanies DC activation to support the anabolic requirements of DC function. We show that early glycolytic activation is a common program for both strong and weak stimuli, but that weakly activated DCs lack long-term HIF-1α-dependent glycolytic reprogramming and retain mitochondrial oxidative metabolism. Early induction of glycolysis is associated with activation of AKT, TBK, and mTOR, and sustained activation of these pathways is associated with long-term glycolytic reprogramming. We show that inhibition of glycolysis impaired maintenance of elongated cell shape, DC motility, CCR7 oligomerization, and DC migration to draining lymph nodes. Together, our results indicate that early induction of glycolysis occurs independent of pro-inflammatory phenotype, and that glycolysis supports DC migratory ability regardless of mitochondrial bioenergetics.
Collapse
|
49
|
Armada‐Moreira A, Thingholm B, Andreassen K, Sebastião AM, Vaz SH, Städler B. On the Assembly of Microreactors with Parallel Enzymatic Pathways. ACTA ACUST UNITED AC 2018; 2:e1700244. [DOI: 10.1002/adbi.201700244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/29/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Adam Armada‐Moreira
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
- Instituto de Farmacologia e Neurociências Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
- Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
| | - Bo Thingholm
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Kristine Andreassen
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
- Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
| | - Sandra H. Vaz
- Instituto de Farmacologia e Neurociências Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
- Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa 1649‐028 Lisboa Portugal
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| |
Collapse
|
50
|
Uhrich D, von Langermann J. Preparation and Characterization of Enzyme Compartments in UV-Cured Polyurethane-Based Materials and Their Application in Enzymatic Reactions. Front Microbiol 2017; 8:2111. [PMID: 29170654 PMCID: PMC5684114 DOI: 10.3389/fmicb.2017.02111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023] Open
Abstract
The preparation and characterization of UV-cured polyurethane-based materials for the mild inclusion immobilization of enzymes was investigated. Full curing of the polymer precursor/enzyme solution mixture was realized by a short irradiation with UV-light at ambient temperatures. The included aqueous enzyme solution remains highly dispersed in the polymer material with an even size distribution throughout the polymer material. The presented concept provides stable enzyme compartments which were applied for an alcohol dehydrogenase-catalyzed reduction reaction in organic solvents. Cofactor regeneration was achieved by a substrate-coupled approach via 2-propanol or an enzyme-coupled approach by a glucose dehydrogenase. This reaction concept can also be used for a simultaneous application of contrary biocatalytic reaction conditions within an enzymatic cascade reaction. Independent polymer-based reaction compartments were provided for two incompatible enzymatic reaction systems (alcohol dehydrogenase and hydroxynitrile lyase), while the relevant reactants diffuse between the applied compartments.
Collapse
Affiliation(s)
- Diana Uhrich
- Biocatalysis Group, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Jan von Langermann
- Biocatalysis Group, Institute of Chemistry, University of Rostock, Rostock, Germany
| |
Collapse
|