451
|
Heterogeneity and deformation behavior of lipid vesicles. Curr Opin Colloid Interface Sci 2022. [DOI: 10.1016/j.cocis.2022.101646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
452
|
Cholesterol-induced robust Ca oscillation in astrocytes required for survival and lipid droplet formation in high-cholesterol condition. iScience 2022; 25:105138. [PMID: 36185358 PMCID: PMC9523397 DOI: 10.1016/j.isci.2022.105138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/08/2022] [Accepted: 09/10/2022] [Indexed: 11/22/2022] Open
Abstract
Cholesterol, one of the major cell membrane components, stabilizes membrane fluidity and regulates signal transduction. Beside its canonical roles, cholesterol has been reported to directly activate signaling pathways such as hedgehog (Hh). We recently found that astrocytes, one of the glial cells, respond to Hh pathway stimulation by Ca signaling. These notions led us to test if extracellularly applied cholesterol triggers Ca signaling in astrocytes. Here, we found that cholesterol application induces robust Ca oscillation only in astrocytes with different properties from the Hh-induced Ca response. The Ca oscillation has a long delay which corresponds to the onset of cholesterol accumulation in the plasma membrane. Blockade of the Ca oscillation resulted in enhancement of astrocytic cell death and disturbance of lipid droplet formation, implying a possibility that the cholesterol-induced Ca oscillation plays important roles in astrocytic survival and cholesterol handling under pathological conditions of cholesterol load such as demyelination. Robust Ca oscillation by cholesterol in astrocytes but not in neurons and microglia Cholesterol-induced Ca oscillation relates to membrane cholesterol accumulation The Ca oscillation is driven via the PLC-IP3 signaling pathway Ca oscillation inhibition leads to astrocytic death and lipid droplet malformation
Collapse
|
453
|
Critical Sites on Ostreolysin Are Responsible for Interaction with Cytoskeletal Proteins. Biomedicines 2022; 10:biomedicines10102442. [PMID: 36289704 PMCID: PMC9598724 DOI: 10.3390/biomedicines10102442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
We explored the structural features of recombinant ostreolysin A (rOlyA), a protein produced by Pleurotus ostreatus and responsible for binding to α/β-tubulin. We found that rOlyA cell internalization is essential for the induction of adipocyte-associated activity, which is mediated by the interaction of rOlyA and microtubule proteins. We created different point mutations at conserved tryptophan (W) sites in rOlyA and analyzed their biological activity in HIB-1B preadipocytes. We demonstrated that the protein’s cell-internalization ability and the differentiated phenotype induced, such as small lipid-droplet formation and gene expression of mitogenesis activity, were impaired in point-mutated proteins W96A and W28A, where W was converted to alanine (A). We also showed that an rOlyA homologue, OlyA6 complexed with mCherry, cannot bind to β-tubulin and does not induce mitochondrial biosynthesis-associated markers, suggesting that the OlyA6 region masked by mCherry is involved in β-tubulin binding. Protein–protein docking simulations were carried out to investigate the binding mode of rOlyA with β-tubulin. Taken together, we identified functional sites in rOlyA that are essential for its binding to β-tubulin and its adipocyte-associated biological activity.
Collapse
|
454
|
Tang R, Liang J, Jing X, Liu T. Discrepancy in Sterol Usage between Two Polyphagous Caterpillars, Mythimna separata and Spodoptera frugiperda. INSECTS 2022; 13:876. [PMID: 36292826 PMCID: PMC9604351 DOI: 10.3390/insects13100876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Insects are sterol auxotrophs and typically obtain sterols from food. However, the sterol demand and metabolic capacity vary greatly among species, even for closely related species. The low survival of many insects on atypical sterols, such as cholestanol and cholestanone, raises the possibility of using sterol-modified plants to control insect herbivore pests. In this study, we evaluated two devastating migratory crop pests, Mythimna separata and Spodoptera frugiperda, in response to atypical sterols and explored the reasons that caused the divergences in sterol nutritional biology between them. Contrary to M. separata, S. frugiperda had unexpectedly high survival on cholestanone, and nearly 80% of the individuals pupated. Comparative studies, including insect response to multiple diets and larval body sterol/steroids analysis, were performed to explain their differences in cholestanone usage. Our results showed that, in comparison to M. separata, the superiority of S. frugiperda on cholestanone can be attributed to its higher efficiency of converting ketone into available stanol and its lower demand for sterols, which resulted in a better survival when cholesterol was unavailable. This research will help us to better understand insect sterol nutritional biology and the potential of using atypical sterols to control herbivorous insect pests.
Collapse
Affiliation(s)
| | | | - Xiangfeng Jing
- Correspondence: (X.J.); (T.L.); Tel.: +86-18220806257 (X.J.); +86-29-87092663 (T.L.)
| | - Tongxian Liu
- Correspondence: (X.J.); (T.L.); Tel.: +86-18220806257 (X.J.); +86-29-87092663 (T.L.)
| |
Collapse
|
455
|
Aparin IO, Yan R, Pelletier R, Choi AA, Danylchuk DI, Xu K, Klymchenko AS. Fluorogenic Dimers as Bright Switchable Probes for Enhanced Super-Resolution Imaging of Cell Membranes. J Am Chem Soc 2022; 144:18043-18053. [PMID: 36153973 DOI: 10.1021/jacs.2c07542] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Super-resolution fluorescence imaging based on single-molecule localization microscopy (SMLM) enables visualizing cellular structures with nanometric precision. However, its spatial and temporal resolution largely relies on the brightness of ON/OFF switchable fluorescent dyes. Moreover, in cell plasma membranes, the single-molecule localization is hampered by the fast lateral diffusion of membrane probes. Here, to address these two fundamental problems, we propose a concept of ON/OFF switchable probes for SMLM (points accumulation for imaging in nanoscale topography, PAINT) based on fluorogenic dimers of bright cyanine dyes. In these probes, the two cyanine units connected with a linker were modified at their extremities with low-affinity membrane anchors. Being self-quenched in water due to intramolecular dye H-aggregation, they displayed light up on reversible binding to lipid membranes. The charged group in the linker further decreased the probe affinity to the lipid membranes, thus accelerating its dynamic reversible ON/OFF switching. The concept was validated on cyanines 3 and 5. SMLM of live cells revealed that the new probes provided higher brightness and ∼10-fold slower diffusion at the cell surface, compared to reference probes Nile Red and DiD, which boosted axial localization precision >3-fold down to 31 nm. The new probe allowed unprecedented observation of nanoscale fibrous protrusions on plasma membranes of live cells with 40 s time resolution, revealing their fast dynamics. Thus, going beyond the brightness limit of single switchable dyes by cooperative dequenching in fluorogenic dimers and slowing down probe diffusion in biomembranes open the route to significant enhancement of super-resolution fluorescence microscopy of live cells.
Collapse
Affiliation(s)
- Ilya O Aparin
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720, United States.,Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| | - Rémi Pelletier
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| | - Alexander A Choi
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720, United States.,Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| | - Dmytro I Danylchuk
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720, United States.,Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 Route du Rhin, 67401 Illkirch, France
| |
Collapse
|
456
|
Zhou Y, Xiao Z, Zhu W. The roles of small extracellular vesicles as prognostic biomarkers and treatment approaches in triple-negative breast cancer. Front Oncol 2022; 12:998964. [PMID: 36212432 PMCID: PMC9537600 DOI: 10.3389/fonc.2022.998964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a particularly aggressive and invasive breast cancer subtype and is associated with poor clinical outcomes. Treatment approaches for TNBC remain limited partly due to the lack of expression of well-known molecular targets. Small extracellular vesicles (sEVs) carrying a variety of bioactive contents play an important role in intercellular communications. The biomolecules including nucleic acids, proteins, and metabolites can be transferred locally or systematically to recipient cells and regulate their biological states and are involved in physiological and pathological processes. Recently, despite the extensive attraction to the physiological functions of sEVs, few studies focus on the roles of sEVs in TNBC. In this review, we will summarize the involvement of sEVs in the tumor microenvironment of TNBC. Moreover, we will discuss the potential roles of sEVs as diagnostic markers and treatment therapy in this heterogeneous breast cancer subtype. We finally summarize the clinical application of sEVs in TNBC.
Collapse
Affiliation(s)
- Yueyuan Zhou
- Department of Clinical Medical Engineering, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- *Correspondence: Yueyuan Zhou,
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wei Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
457
|
Oğul Y, Gür F, Gür B, Cengiz M, Sarı RA, Kızıltunç A. Decreased Na +/K + pump activity in the erythrocyte membrane due to malondialdehyde in rheumatoid arthritis: an in vivo and in silico study. Can J Physiol Pharmacol 2022; 100:968-982. [PMID: 36148907 DOI: 10.1139/cjpp-2022-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Apart from demonstrating the interaction behavior of malondialdehyde (MDA) with Na+/K+-ATPase using in silico, the current study aims to investigate the effect of rheumatoid arthritis-related oxidative stress on Na+/K+-ATPase activity that is present in the erythrocyte cell membrane, which is rich in proteins vulnerable to damage from MDA and other free radicals. The target population of this study consists of 28 rheumatoid arthritis patients and 20 healthy volunteers whose MDA levels and Na+/K+-ATPase activity were determined. It was shown that MDA levels of rheumatoid arthritis patients increased (p < 0.001) and their Na+/K+-ATPase activity noticeably decreased when compared to those of healthy individuals. Also, according to this in silico modeling, MDA decreased Na+/K+-ATPase activity in line with the correlation analyses. Consequently, while elevated levels of MDA in the rheumatoid arthritis group were suggestive of oxidative stress, a decreased Na+/K+-ATPase-activity led us to speculate that the cellular membrane had sustained injury. Therefore, our results could be useful in explaining how MDA affects Na+/K+-ATPase activity in the interior of a specific molecular pathway.
Collapse
Affiliation(s)
- Yasemin Oğul
- Department of Medicinal Biochemistry, Regional Training and Research Hospital, Health Sciences University, Erzurum, 25240, Turkey
| | - Fatma Gür
- Department of Medical Services and Techniques, Health Services Vocational School, Ataturk University, Erzurum, 25240, Turkey
| | - Bahri Gür
- Department of Biochemistry, Faculty of Sciences and Arts, Iğdır University, Iğdır, 76000, Turkey
| | - Mustafa Cengiz
- Department of Elementary Education, Faculty of Education, Siirt University, 56100 Siirt, Turkey
| | - Refik Ali Sarı
- Department of Internal Diseases, Faculty of Medicine, Karadeniz Technical University, Trabzon, 61080, Turkey
| | - Ahmet Kızıltunç
- Department of Biochemistry, Faculty of Medicine, Ataturk University, Erzurum, 25240, Turkey
| |
Collapse
|
458
|
Tsioulos G, Grigoropoulos I, Moschopoulos CD, Shapira S, Poulakou G, Antoniadou A, Boumpas D, Arber N, Tsiodras S. Insights into CD24 and Exosome Physiology and Potential Role in View of Recent Advances in COVID-19 Therapeutics: A Narrative Review. Life (Basel) 2022; 12:1472. [PMID: 36294907 PMCID: PMC9604962 DOI: 10.3390/life12101472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/17/2022] [Indexed: 08/30/2023] Open
Abstract
Cluster of differentiation (CD) 24, a long-known protein with multifaceted functions, has gained attention as a possible treatment for Coronavirus Disease 19 (COVID-19) due to its known anti-inflammatory action. Extracellular vesicles (EVs), such as exosomes and microvesicles, may serve as candidate drug delivery platforms for novel therapeutic approaches in COVID-19 and various other diseases due to their unique characteristics. In the current review, we describe the physiology of CD24 and EVs and try to elucidate their role, both independently and as a combination, in COVID-19 therapeutics. CD24 may act as an important immune regulator in diseases with complex physiologies characterized by excessive inflammation. Very recent data outline a possible therapeutic role not only in COVID-19 but also in other similar disease states, e.g., acute respiratory distress syndrome (ARDS) and sepsis where immune dysregulation plays a key pathophysiologic role. On the other hand, CD24, as well as other therapeutic molecules, can be administered with the use of exosomes, exploiting their unique characteristics to create a novel drug delivery platform as outlined in recent clinical efforts. The implications for human therapeutics in general are huge with regard to pharmacodynamics, pharmacokinetics, safety, and efficacy that will be further elucidated in future randomized controlled trials (RCTs).
Collapse
Affiliation(s)
- Georgios Tsioulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Charalampos D. Moschopoulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Shiran Shapira
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Garyfallia Poulakou
- 3rd Department of Internal Medicine, Medical School, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dimitrios Boumpas
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
459
|
Li L, Hu J, Różycki B, Ji J, Song F. Interplay of receptor-ligand binding and lipid domain formation during cell adhesion. Front Mol Biosci 2022; 9:1019477. [PMID: 36203878 PMCID: PMC9531914 DOI: 10.3389/fmolb.2022.1019477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022] Open
Abstract
Cell adhesion involved in biological processes such as cell migration, immune responses, and cancer metastasis, is mediated by the specific binding of receptor and ligand proteins. Some of these proteins exhibit affinity for nanoscale lipid clusters in cell membranes. A key question is how these nanoscale lipid clusters influence and react to the receptor-ligand binding during cell adhesion. In this article, we review recent computational studies that shed new light on the interplay of the receptor-ligand binding and the formation of lipid domains in adhering membranes. These studies indicate that the receptor-ligand binding promotes coalescence of lipid clusters into mesoscale domains, which, in turn, enhances both the affinity and cooperativity of the receptor-ligand binding in cell-cell adhesion with mobile ligands. In contrast, in the case of cell-extracellular matrix adhesion with immobile ligands, the receptor-ligand binding and the lipid cluster coalescence can be correlated or anti-correlated, depending strongly on the ligand distribution. These findings deepen our understanding of correlations between cell adhesion and membrane heterogeneities.
Collapse
Affiliation(s)
- Long Li
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Jinglei Hu
- Kuang Yaming Honors School and Institute for Brain Sciences, Nanjing University, Nanjing, China
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Bartosz Różycki
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Jing Ji
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Jinglei Hu, ; Bartosz Różycki, ; Jing Ji,
| | - Fan Song
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
460
|
Liu Z, Liu Y, Xing T, Li J, Zhang L, Jiang Y, Gao F. Transcriptome analysis reveals the mechanism of chronic heat stress on meat quality of broilers. J Anim Sci Biotechnol 2022; 13:110. [PMID: 36117193 PMCID: PMC9484139 DOI: 10.1186/s40104-022-00759-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/13/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chronic heat stress has a negative impact on poultry meat quality. Although this has been extensively investigated, previous studies have primarily focused on metabolic alterations and oxidative stress in the pectoralis major (PM) muscle under chronic heat stress, and not all of the underlying molecular mechanisms are completely understood.
Methods
A total of 144 male Arbor Acres broilers (28 d old) were randomly allocated into 3 treatment groups: (1) the normal control (NC) group, with broilers raised at 22 °C and fed a basal diet; (2) the heat stress (HS) group, with birds raised at 32 °C and fed a basal diet; and (3) the pair-fed (PF) group, with birds raised at 22 °C and fed the amount of feed equal to the feed consumed on the previous day by the HS group. The experiment lasted for 14 d.
Results
Chronic heat stress decreased the average daily feed intake and average daily gain, increased feed:gain ratio (P < 0.05); and increased drip loss, cooking loss, shear force, hardness, and decreased pH, redness (a*); and springiness of PM muscle (P < 0.05). Furthermore, chronic heat stress decreased muscle fiber density, increased connective tissue, and led to intracellular vacuolation. The transcriptome analyses indicated that the effect of chronic heat stress on meat quality was not only related to metabolism and oxidative stress, but also to signal transduction, immune system, transport and catabolism, cell growth and death, and muscle structure.
Conclusions
Chronic heat stress has a negative impact on the growth performance, meat quality, and the PM muscle structure of broilers. Transcriptome analysis revealed a comprehensive understanding of the mechanism of the chronic heat stress-induced deterioration of broiler meat quality at the transcriptional level.
Collapse
|
461
|
Mohammed SA, Saini RV, Jha AK, Hadda V, Singh AK, Prakash H. Sphingolipids, mycobacteria and host: Unraveling the tug of war. Front Immunol 2022; 13:1003384. [PMID: 36189241 PMCID: PMC9521350 DOI: 10.3389/fimmu.2022.1003384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shakeel Ahmed Mohammed
- Amity Institute of Virology and Immunology, Amity University, Noida, India
- Department of Biotechnology, Maharishi Markandeshwar (M. M). Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Reena Vohra Saini
- Department of Biotechnology, Maharishi Markandeshwar (M. M). Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | | | - Vijay Hadda
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Kumar Singh
- Experimental Animal Facility, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj Agra, India
| | - Hridayesh Prakash
- Department of Biotechnology, Maharishi Markandeshwar (M. M). Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala, India
- *Correspondence: Hridayesh Prakash,
| |
Collapse
|
462
|
Atreya KB, Saba JD. Neurological Consequences of Sphingosine Phosphate Lyase Insufficiency. Front Cell Neurosci 2022; 16:938693. [PMID: 36187293 PMCID: PMC9519528 DOI: 10.3389/fncel.2022.938693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In 2017, an inborn error of metabolism caused by recessive mutations in SGPL1 was discovered. The disease features steroid-resistant nephrotic syndrome, adrenal insufficiency, and neurological defects. The latter can include sensorineural hearing loss, cranial nerve defects, peripheral neuropathy, abnormal brain development, seizures and/or neurodegeneration. SGPL1 encodes the pyridoxal-5’-phosphate (PLP) dependent enzyme sphingosine phosphate lyase (SPL), and the condition is now referred to as SPL insufficiency syndrome (SPLIS). SPL catalyzes the final step in the degradative pathway of sphingolipids in which the bioactive sphingolipid sphingosine-1-phosphate (S1P) is irreversibly degraded to a long chain aldehyde and phosphoethanolamine (PE). SPL guards the only exit point for sphingolipid metabolism, and its inactivation leads to accumulation of various types of sphingolipids which have biophysical roles in plasma membrane rafts and myelin, and signaling roles in cell cycle progression, vesicular trafficking, cell migration, and programmed cell death. In addition, the products of the SPL reaction have biological functions including regulation of autophagic flux, which is important in axonal and neuronal integrity. In this review, the neurological manifestations of SPLIS will be described, and insights regarding the neurological consequences of SPL insufficiency from the study of brain-specific SPL knockout mice and Drosophila SPL mutants will be summarized.
Collapse
Affiliation(s)
- Krishan B. Atreya
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Julie D. Saba
- Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Julie D. Saba
| |
Collapse
|
463
|
Hydroxychloroquine blocks SARS-CoV-2 entry into the endocytic pathway in mammalian cell culture. Commun Biol 2022; 5:958. [PMID: 36104427 PMCID: PMC9472185 DOI: 10.1038/s42003-022-03841-8] [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] [Received: 11/08/2021] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Hydroxychloroquine (HCQ), a drug used to treat lupus and malaria, was proposed as a treatment for SARS-coronavirus-2 (SARS-CoV-2) infection, albeit with controversy. In vitro, HCQ effectively inhibits viral entry, but its use in the clinic has been hampered by conflicting results. A better understanding of HCQ’s mechanism of actions in vitro is needed. Recently, anesthetics were shown to disrupt ordered clusters of monosialotetrahexosylganglioside1 (GM1) lipid. These same lipid clusters recruit the SARS-CoV-2 surface receptor angiotensin converting enzyme 2 (ACE2) to endocytic lipids, away from phosphatidylinositol 4,5 bisphosphate (PIP2) clusters. Here we employed super-resolution imaging of cultured mammalian cells (VeroE6, A549, H1793, and HEK293T) to show HCQ directly perturbs clustering of ACE2 receptor with both endocytic lipids and PIP2 clusters. In elevated (high) cholesterol, HCQ moves ACE2 nanoscopic distances away from endocytic lipids. In cells with resting (low) cholesterol, ACE2 primarily associates with PIP2 clusters, and HCQ moves ACE2 away from PIP2 clusters—erythromycin has a similar effect. We conclude HCQ inhibits viral entry through two distinct mechanisms in high and low tissue cholesterol and does so prior to inhibiting cathepsin-L. HCQ clinical trials and animal studies will need to account for tissue cholesterol levels when evaluating dosing and efficacy. Super-resolution microscopy in cultured cells is employed to dissect the effect of hydroxychloroquine (HCQ) at the plasma membrane and HCQ directly perturbs clustering of the SARS-CoV-2 receptor ACE2 with endocytic lipids and PIP2 clusters.
Collapse
|
464
|
Hartel JC, Merz N, Grösch S. How sphingolipids affect T cells in the resolution of inflammation. Front Pharmacol 2022; 13:1002915. [PMID: 36176439 PMCID: PMC9513432 DOI: 10.3389/fphar.2022.1002915] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of proper resolution of inflammation rather than counteracting it, gained a lot of attention in the past few years. Re-assembly of tissue and cell homeostasis as well as establishment of adaptive immunity after inflammatory processes are the key events of resolution. Neutrophiles and macrophages are well described as promotors of resolution, but the role of T cells is poorly reviewed. It is also broadly known that sphingolipids and their imbalance influence membrane fluidity and cell signalling pathways resulting in inflammation associated diseases like inflammatory bowel disease (IBD), atherosclerosis or diabetes. In this review we highlight the role of sphingolipids in T cells in the context of resolution of inflammation to create an insight into new possible therapeutical approaches.
Collapse
Affiliation(s)
- Jennifer Christina Hartel
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
- Department of Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany
| | - Nadine Merz
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- *Correspondence: Sabine Grösch,
| |
Collapse
|
465
|
Meléndez AV, Velasco Cárdenas RMH, Lagies S, Strietz J, Siukstaite L, Thomas OS, Tomisch J, Weber W, Kammerer B, Römer W, Minguet S. Novel lectin-based chimeric antigen receptors target Gb3-positive tumour cells. Cell Mol Life Sci 2022; 79:513. [PMID: 36097202 PMCID: PMC9468074 DOI: 10.1007/s00018-022-04524-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 07/19/2022] [Accepted: 07/31/2022] [Indexed: 11/05/2022]
Abstract
The link between cancer and aberrant glycosylation has recently become evident. Glycans and their altered forms, known as tumour-associated carbohydrate antigens (TACAs), are diverse, complex and difficult to target therapeutically. Lectins are naturally occurring glycan-binding proteins that offer a unique opportunity to recognise TACAs. T cells expressing chimeric antigen receptors (CARs) have proven to be a successful immunotherapy against leukaemias, but so far have shown limited success in solid tumours. We developed a panel of lectin-CARs that recognise the glycosphingolipid globotriaosylceramide (Gb3), which is overexpressed in various cancers, such as Burkitt's lymphoma, colorectal, breast and pancreatic. We have selected the following lectins: Shiga toxin's B-subunit from Shigella dysenteriae, LecA from Pseudomonas aeruginosa, and the engineered lectin Mitsuba from Mytilus galloprovincialis as antigen-binding domains and fused them to a well-known second-generation CAR. The Gb3-binding lectin-CARs have demonstrated target-specific cytotoxicity against Burkitt's lymphoma-derived cell lines as well as solid tumour cells from colorectal and triple-negative breast cancer. Our findings reveal the big potential of lectin-based CARs as therapeutical applications to target Gb3 and other TACAs expressed in haematological malignancies and solid tumours.
Collapse
Affiliation(s)
- Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Rubí M-H Velasco Cárdenas
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Simon Lagies
- Institute of Organic Chemistry, Albert-Ludwigs-University Freiburg, Albertstraße 21, 79102, Freiburg, Germany
| | | | - Lina Siukstaite
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Oliver S Thomas
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Jana Tomisch
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Wilfried Weber
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany
| | - Bernd Kammerer
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Institute of Organic Chemistry, Albert-Ludwigs-University Freiburg, Albertstraße 21, 79102, Freiburg, Germany
- Centre for Integrative Signalling Analysis, University of Freiburg, Habsburgerstraße 49, 79104, Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
- Center of Chronic Immunodeficiency (CCI), University Clinics and Medical Faculty, Freiburg, Germany.
| |
Collapse
|
466
|
Himbert S, Rheinstädter MC. Structural and mechanical properties of the red blood cell's cytoplasmic membrane seen through the lens of biophysics. Front Physiol 2022; 13:953257. [PMID: 36171967 PMCID: PMC9510598 DOI: 10.3389/fphys.2022.953257] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/15/2022] [Indexed: 11/27/2022] Open
Abstract
Red blood cells (RBCs) are the most abundant cell type in the human body and critical suppliers of oxygen. The cells are characterized by a simple structure with no internal organelles. Their two-layered outer shell is composed of a cytoplasmic membrane (RBC cm ) tethered to a spectrin cytoskeleton allowing the cell to be both flexible yet resistant against shear stress. These mechanical properties are intrinsically linked to the molecular composition and organization of their shell. The cytoplasmic membrane is expected to dominate the elastic behavior on small, nanometer length scales, which are most relevant for cellular processes that take place between the fibrils of the cytoskeleton. Several pathologies have been linked to structural and compositional changes within the RBC cm and the cell's mechanical properties. We review current findings in terms of RBC lipidomics, lipid organization and elastic properties with a focus on biophysical techniques, such as X-ray and neutron scattering, and Molecular Dynamics simulations, and their biological relevance. In our current understanding, the RBC cm 's structure is patchy, with nanometer sized liquid ordered and disordered lipid, and peptide domains. At the same time, it is surprisingly soft, with bending rigidities κ of 2-4 kBT. This is in strong contrast to the current belief that a high concentration of cholesterol results in stiff membranes. This extreme softness is likely the result of an interaction between polyunsaturated lipids and cholesterol, which may also occur in other biological membranes. There is strong evidence in the literature that there is no length scale dependence of κ of whole RBCs.
Collapse
Affiliation(s)
- Sebastian Himbert
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| | - Maikel C. Rheinstädter
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Origins Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
467
|
Wei T, Zhang L, Zhang Y. Cholesterol Distribution in Small Unilamellar Vesicles. J Phys Chem B 2022; 126:7135-7142. [PMID: 36074983 DOI: 10.1021/acs.jpcb.2c01785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The plasma membrane has heterogeneity, where the constituents are not only asymmetrically distributed between the inner leaflet and the outer leaflet but also laterally organized within each leaflet. There is still an ongoing controversy over the cholesterol distribution between the two leaflets, and it is also significant to explore the lateral flow and localization of cholesterol. The unilamellar vesicle is extensively employed as a simplified model of the plasma membrane in research studies. In this work, we study the spontaneous spatial distribution of cholesterol in the small unilamellar vesicles with a single type of phospholipid as the bilayer backbone through coarse-grained molecular dynamics simulations. The results show that in a spherical vesicle under ambient pressure, cholesterol is more abundant in the inner leaflet than in the outer leaflet. As the vesicle is deformed under pressurization, the net lateral flow of cholesterol in the two leaflets is in the exactly opposite directions, finally leading to a distribution strongly associated with the monolayer curvature. One of the possible explanations for our results is from the point of view of curvature elastic energy. Another possibility is from the point of view of stress: according to the correlation between the cholesterol distribution and the tail angles of lipids, it is suggested that the possible governing mechanism for the distribution of cholesterol in a membrane is to alleviate the mismatch of stress between the two leaflets. Additionally, we obtain the effect of cholesterol infiltration on the bending modulus and the spontaneous curvature of the vesicles.
Collapse
Affiliation(s)
- Tanlin Wei
- School of Physics, Sun Yat-Sen University, Guangzhou 510275, China
| | - Lei Zhang
- School of Physics, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yong Zhang
- School of Physics, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
468
|
Kotani N, Araki T, Miyagawa-Yamaguchi A, Amimoto T, Nakano M, Honke K. Proximity Labeling and Proteomics: Get to Know Neighbors. Methods Enzymol 2022; 679:131-162. [PMID: 36682860 DOI: 10.1016/bs.mie.2022.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Protein-protein interactions are essential in biological reactions and fundamental to cell-cell communication (e.g., the binding of secreted proteins, such as hormones, to cell membrane receptors) and the subsequent intracellular signal transduction cascade. Several studies have been extensively carried out on protein-protein interactions because they have the potential to resolve various problems in molecular biology. Biochemical methods, such as chemical cross-linking and immunoprecipitation, have long been used to analyze which proteins interact with each other. However, there are some problems, such as unphysiological states and non-specific binding, that require the development of more useful experimental methods. This chapter discusses the "proximity labeling (Proteomics)" analysis technique, which has been attracting attention in protein-protein interaction analysis in recent years and is used in many biological studies. "Membrane proximity labeling (proteomics)," which analyzes the interaction of cell membrane proteins, and "intracellular proximity labeling (proteomics)" will be explained in-depth.
Collapse
Affiliation(s)
- Norihiro Kotani
- Medical Research Center, Saitama Medical University, Saitama, Japan; Department of Biochemistry, Saitama Medical University, Saitama, Japan.
| | - Tomoyuki Araki
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | | | - Tomoko Amimoto
- Natural Science Center for Basic Research and Development, Hiroshima University, Higashi-Hiroshima, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Koichi Honke
- Department of Biochemistry, Kochi University Medical School, Nankoku, Japan
| |
Collapse
|
469
|
Wirth D, Paul MD, Pasquale EB, Hristova K. Direct quantification of ligand-induced lipid and protein microdomains with distinctive signaling properties. CHEMSYSTEMSCHEM 2022; 4:e202200011. [PMID: 36337751 PMCID: PMC9634703 DOI: 10.1002/syst.202200011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 11/08/2022]
Abstract
Lipid rafts are ordered lipid domains that are enriched in saturated lipids, such as the ganglioside GM1. While lipid rafts are believed to exist in cells and to serve as signaling platforms through their enrichment in signaling components, they have not been directly observed in the plasma membrane without treatments that artificially cluster GM1 into large lattices. Here, we report that microscopic GM1-enriched domains can form, in the plasma membrane of live mammalian cells expressing the EphA2 receptor tyrosine kinase in response to its ligand ephrinA1-Fc. The GM1-enriched microdomains form concomitantly with EphA2-enriched microdomains. To gain insight into how plasma membrane heterogeneity controls signaling, we quantify the degree of EphA2 segregation and study initial EphA2 signaling steps in both EphA2-enriched and EphA2-depleted domains. By measuring dissociation constants, we demonstrate that the propensity of EphA2 to oligomerize is similar in EphA2-enriched and -depleted domains. However, surprisingly, EphA2 interacts preferentially with its downstream effector SRC in EphA2-depleted domains. The ability to induce microscopic GM1-enriched domains in live cells using a ligand for a transmembrane receptor will give us unprecedented opportunities to study the biophysical chemistry of lipid rafts.
Collapse
Affiliation(s)
- Daniel Wirth
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218
| | - Michael D. Paul
- Program in Molecular Biophysics, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218
| | - Elena B. Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Road, La Jolla, CA 92037
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218
- Program in Molecular Biophysics, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218
| |
Collapse
|
470
|
Karpouzas GA, Papotti B, Ormseth S, Palumbo M, Hernandez E, Adorni MP, Zimetti F, Budoff M, Ronda N. Serum cholesterol loading capacity on macrophages is linked to coronary atherosclerosis and cardiovascular event risk in rheumatoid arthritis. RMD Open 2022; 8:rmdopen-2022-002411. [PMID: 36113961 PMCID: PMC9486392 DOI: 10.1136/rmdopen-2022-002411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/03/2022] [Indexed: 11/05/2022] Open
Abstract
Objectives Cholesterol loading capacity (CLC) describes the ability of serum to deliver cholesterol to cells. It is linked to foam cell formation, a pivotal step in atherosclerotic plaque development. We evaluate the associations of CLC with coronary atherosclerosis presence, burden and cardiovascular risk in patients with rheumatoid arthritis (RA). Methods Coronary atherosclerosis (any, high-risk low-attenuation plaque and obstructive plaque) was evaluated with CT angiography in 141 patients. Participants were prospectively followed for 6.0±2.4 years and cardiovascular events including cardiac death, myocardial infarction, unstable angina, stroke, claudication, revascularisation and hospitalised heart failure were recorded. CLC was quantified as intracellular cholesterol in human macrophages after incubation with patient serum. Results CLC was not linked to overall plaque presence or burden after adjustments for atherosclerotic cardiovascular disease (ASCVD) score, statin use and low-density lipoprotein cholesterol. However, CLC associated with presence and numbers of any, low-attenuation and obstructive plaques exclusively in biologic disease-modifying antirheumatic drugs (bDMARD) non-users (p for interaction ≤0.018). CLC associated with cardiovascular event risk overall after adjustments for ASCVD and number of segments with plaque (HR=1.76 (95% CI 1.16 to 2.67) per 1 SD increase in CLC, p=0.008). Additionally, bDMARD use modified the impact of CLC on event risk; CLC associated with events in bDMARD non-users (HR=2.52 (95% CI 1.36 to 4.65) per 1SD increase in CLC, p=0.003) but not users. Conclusion CLC was linked to long-term cardiovascular event risk in RA and associated with high-risk low attenuation and obstructive coronary plaque presence and burden in bDMARD non-users. Its prospective validation as a predictive biomarker may be, therefore, warranted.
Collapse
Affiliation(s)
- George Athanasios Karpouzas
- Internal Medicine-Rheumatology, Lundquist Institute, Torrance, California, USA .,Department of Rheumatology, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Sarah Ormseth
- Internal Medicine-Rheumatology, Lundquist Institute, Torrance, California, USA
| | | | | | | | - Francesca Zimetti
- Department of Pharmacy, University of Parma, Parma, Emilia-Romagna, Italy
| | - Matthew Budoff
- Internal Medicine, Lundquist Institute, Torrance, California, USA
| | - Nicoletta Ronda
- Department of Pharmacy, University of Parma, Parma, Emilia-Romagna, Italy
| |
Collapse
|
471
|
Varghese PM, Kishore U, Rajkumari R. Innate and adaptive immune responses against Influenza A Virus: Immune evasion and vaccination strategies. Immunobiology 2022; 227:152279. [DOI: 10.1016/j.imbio.2022.152279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
|
472
|
Wang M, Deng R. Effects of carbon black nanoparticles and high humidity on the lung metabolome in Balb/c mice with established allergic asthma. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:65100-65111. [PMID: 35484453 DOI: 10.1007/s11356-022-20349-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
In respiratory diseases, the induction of allergic asthma has gradually aroused public concerns. Co-exposures of environmental risk factors such as nanoparticles and high humidity could play important roles in the development of allergic asthma. However, the relevant researches are still lacking and the involved mechanisms, especially metabolic changes, remain unclear. We took the lead in studying the combined induction effect and underlying mechanisms of carbon black nanoparticles (CB NPs) and high humidity on allergic asthma. In this work, murine models of allergic asthma were established with ovalbumin under the single and combined exposures of 15 μg/kg CB NPs and 90% relative humidity. The two risk factors, particularly their co-exposure, exhibited adjuvant effect on airway hyperresponsiveness, remodeling, and inflammation in Balb/c mice. Untargeted metabolomics identified the potential biomarkers in lung for asthma occurrence and for asthma exacerbation caused by CB NPs and high humidity. The significantly dysregulated metabolic pathways in asthmatic mice were proposed, and the disturbed metabolic pathways under the exposures of CB NPs and/or high humidity were mainly implicated in asthma symptoms. This work sheds light on the understanding for health risks of NP pollutions and high environmental humidity and contributes to useful biomarker identification and asthma control.
Collapse
Affiliation(s)
- Mingpu Wang
- School of Civil Engineering, Chongqing University, Chongqing, 400045, China
| | - Rui Deng
- School of Civil Engineering, Chongqing University, Chongqing, 400045, China.
| |
Collapse
|
473
|
Allender DW, Schick M. Extent of raft composition in a model plasma membrane. Biophys J 2022:S0006-3495(22)00723-8. [PMID: 36050886 DOI: 10.1016/j.bpj.2022.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 11/29/2022] Open
Abstract
"Rafts" are nanometer-size inhomogeneities in the plasma membrane that, in the outer leaflet, are enriched in sphingomyelin and cholesterol. They are thought to provide a platform for proteins to carry out biological processes (1-4). Here we employ a model asymmetric plasma membrane to address the question of the range of sphingomyelin and cholesterol compositions in which one would expect the formation of rafts. We define a weight for the likelihood of raft formation, and evaluate it as a function of the sphingomyelin mole fraction in the outer leaflet for three bilayers with total cholesterol mole fractions of 0.30, 0.40, and 0.50. Not surprisingly, the weight decreases when there is little sphingomyelin. Less expected, we find that the weight also decreases when there is a large mole fraction of sphingomyelin. The weight is largest in the bilayer with a total cholesterol mole fraction of 0.30, and decreases rapidly with increasing total cholesterol. We explicate the reasons for these behaviors. In the 0.30 cholesterol bilayer, the largest weight occurrs at a sphingomyelin mole fraction in the outer leaflet of approximately 0.23. The weight falls to one half its maximum value at sphingomylin mole fractions of 0.15 and 0.33. In terms of the sphingomyelin mole fraction of the asymmetric bilayer, the maximum weight occurs at 0.12 and falls to half maximum at 0.08 and 0.17.
Collapse
Affiliation(s)
- David W Allender
- Department of Physics, University of Washington, Seattle WA; Department of Physics, Kent State University, Kent OH
| | - Michael Schick
- Department of Physics, University of Washington, Seattle WA
| |
Collapse
|
474
|
Zhang S, Zhu N, Li HF, Gu J, Zhang CJ, Liao DF, Qin L. The lipid rafts in cancer stem cell: a target to eradicate cancer. Stem Cell Res Ther 2022; 13:432. [PMID: 36042526 PMCID: PMC9429646 DOI: 10.1186/s13287-022-03111-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells with stem cell properties that sustain cancers, which may be responsible for cancer metastasis or recurrence. Lipid rafts are cholesterol- and sphingolipid-enriched microdomains in the plasma membrane that mediate various intracellular signaling. The occurrence and progression of cancer are closely related to lipid rafts. Emerging evidence indicates that lipid raft levels are significantly enriched in CSCs compared to cancer cells and that most CSC markers such as CD24, CD44, and CD133 are located in lipid rafts. Furthermore, lipid rafts play an essential role in CSCs, specifically in CSC self-renewal, epithelial-mesenchymal transition, drug resistance, and CSC niche. Therefore, lipid rafts are critical regulatory platforms for CSCs and promising therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hong Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Chan Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Duan Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China. .,Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Hunan University of Chinese Medicine, Changsha, China. .,Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
475
|
Pal A, Gori S, Yoo SW, Thomas AG, Wu Y, Friedman J, Tenora L, Bhasin H, Alt J, Haughey N, Slusher BS, Rais R. Discovery of Orally Bioavailable and Brain-Penetrable Prodrugs of the Potent nSMase2 Inhibitor DPTIP. J Med Chem 2022; 65:11111-11125. [PMID: 35930706 PMCID: PMC9980655 DOI: 10.1021/acs.jmedchem.2c00562] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular vesicles (EVs) can carry pathological cargo and play an active role in disease progression. Neutral sphingomyelinase-2 (nSMase2) is a critical regulator of EV biogenesis, and its inhibition has shown protective effects in multiple disease states. 2,6-Dimethoxy-4-(5-phenyl-4-thiophen-2-yl-1H-imidazol-2-yl)phenol (DPTIP) is one of the most potent (IC50 = 30 nM) inhibitors of nSMase2 discovered to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), limiting its clinical development. To overcome DPTIP's PK limitations, we synthesized a series of prodrugs by masking its phenolic hydroxyl group. When administered orally, the best prodrug (P18) with a 2',6'-diethyl-1,4'-bipiperidinyl promoiety exhibited >fourfold higher plasma (AUC0-t = 1047 pmol·h/mL) and brain exposures (AUC0-t = 247 pmol·h/g) versus DPTIP and a significant enhancement of DPTIP half-life (2 h vs ∼0.5 h). In a mouse model of acute brain injury, DPTIP released from P18 significantly inhibited IL-1β-induced EV release into plasma and attenuated nSMase2 activity. These studies report the discovery of a DPTIP prodrug with potential for clinical translation.
Collapse
Affiliation(s)
- Arindom Pal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sadakatali Gori
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seung-wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jacob Friedman
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Lukáš Tenora
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Harshit Bhasin
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Oncology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Medicine, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| |
Collapse
|
476
|
Rudajev V, Novotny J. Cholesterol as a key player in amyloid β-mediated toxicity in Alzheimer’s disease. Front Mol Neurosci 2022; 15:937056. [PMID: 36090253 PMCID: PMC9453481 DOI: 10.3389/fnmol.2022.937056] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is one of the most devastating and widespread diseases worldwide, mainly affecting the aging population. One of the key factors contributing to AD-related neurotoxicity is the production and aggregation of amyloid β (Aβ). Many studies have shown the ability of Aβ to bind to the cell membrane and disrupt its structure, leading to cell death. Because amyloid damage affects different parts of the brain differently, it seems likely that not only Aβ but also the nature of the membrane interface with which the amyloid interacts, helps determine the final neurotoxic effect. Because cholesterol is the dominant component of the plasma membrane, it plays an important role in Aβ-induced toxicity. Elevated cholesterol levels and their regulation by statins have been shown to be important factors influencing the progression of neurodegeneration. However, data from many studies have shown that cholesterol has both neuroprotective and aggravating effects in relation to the development of AD. In this review, we attempt to summarize recent findings on the role of cholesterol in Aβ toxicity mediated by membrane binding in the pathogenesis of AD and to consider it in the broader context of the lipid composition of cell membranes.
Collapse
|
477
|
Abstract
While it has long been established that cell membranes are complex assemblies of proteins and bilayer-forming lipids, the inherent mobility and wide-ranging heterogeneity of the lipids have limited our ability to understand cell-membrane structure at a molecular level. Consequently, little is yet known about the protein-lipid and lipid-lipid interplay that exists in situ. The present study exploits the regular architecture of a cholinergic cell membrane to determine how the phospholipid and cholesterol organization is influenced by the protein surfaces and by differences in cholesterol concentration between the two leaflets. Lipids in the leaflet containing the most cholesterol form an ordered sterol-hydrocarbon “skin.” This hitherto unobserved hydrophobic-core structure has far-reaching implications in terms of how cholesterol-rich membranes are constructed and function. Cell membranes are complex assemblies of proteins and lipids making transient or long-term associations that have yet to be characterized at a molecular level. Here, cryo-electron microscopy is applied to determine how phospholipids and cholesterol arrange between neighboring proteins (nicotinic acetylcholine receptors) of Torpedo cholinergic membrane. The lipids exhibit distinct properties in the two leaflets of the bilayer, influenced by the protein surfaces and by differences in cholesterol concentration. In the outer leaflet, the lipids show no consistent motif away from the protein surfaces, in keeping with their assumed fluidity. In the inner leaflet, where the cholesterol concentration is higher, the lipids organize into extensive close-packed linear arrays. These arrays are built from the sterol groups of cholesterol and the initial saturated portions of the phospholipid hydrocarbon chains. Together, they create an ordered ∼7 Å-thick “skin” within the hydrophobic core of the bilayer. The packing of lipids in the arrays appears to bear a close relationship to the linear cholesterol arrays that form crystalline monolayers at the air-water interface.
Collapse
|
478
|
Grajeda BI, De Chatterjee A, Villalobos CM, Pence BC, Ellis CC, Enriquez V, Roy S, Roychowdhury S, Neumann AK, Almeida IC, Patterson SE, Das S. Giardial lipid rafts share virulence factors with secreted vesicles and participate in parasitic infection in mice. Front Cell Infect Microbiol 2022; 12:974200. [PMID: 36081774 PMCID: PMC9445159 DOI: 10.3389/fcimb.2022.974200] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Giardia lamblia, a protozoan parasite, is a major cause of waterborne infection, worldwide. While the trophozoite form of this parasite induces pathological symptoms in the gut, the cyst form transmits the infection. Since Giardia is a noninvasive parasite, the actual mechanism by which it causes disease remains elusive. We have previously reported that Giardia assembles cholesterol and GM1 glycosphingolipid-enriched lipid rafts (LRs) that participate in encystation and cyst production. To further delineate the role of LRs in pathogenesis, we isolated LRs from Giardia and subjected them to proteomic analysis. Various cellular proteins including potential virulence factors-e.g., giardins, variant surface proteins, arginine deaminases, elongation factors, ornithine carbomyltransferases, and high cysteine-rich membrane proteins-were found to be present in LRs. Since Giardia secretes virulence factors encapsulated in extracellular vesicles (EVs) that induce proinflammatory responses in hosts, EVs released by the parasite were isolated and subjected to nanoparticle tracking and proteomic analysis. Two types of EV-i.e., small vesicles (SVs; <100 nm, exosome-like particles) and large vesicles (LVs; 100-400 nm, microvesicle-like particles)-were identified and found to contain a diverse group of proteins including above potential virulence factors. Although pretreatment of the parasite with two giardial lipid raft (gLR) disruptors, nystatin (27 μM) and oseltamivir (20 μM), altered the expression profiles of virulence factors in LVs and SVs, the effects were more robust in the case of SVs. To examine the potential role of rafts and vesicles in pathogenicity, Giardia-infected mice were treated with oseltamivir (1.5 and 3.0 mg/kg), and the shedding of cysts were monitored. We observed that this drug significantly reduced the parasite load in mice. Taken together, our results suggest that virulence factors partitioning in gLRs, released into the extracellular milieu via SVs and LVs, participate in spread of giardiasis and could be targeted for future drug development.
Collapse
Affiliation(s)
- Brian I. Grajeda
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Atasi De Chatterjee
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Carmen M. Villalobos
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Breanna C. Pence
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Cameron C. Ellis
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Vanessa Enriquez
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sourav Roy
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sukla Roychowdhury
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Aaron K. Neumann
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Igor C. Almeida
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Steven E. Patterson
- Center for Drug Design, University of Minnesota, Minneapolis, MN, United States
| | - Siddhartha Das
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
479
|
AIEgen-Peptide Bioprobes for the Imaging of Organelles. BIOSENSORS 2022; 12:bios12080667. [PMID: 36005064 PMCID: PMC9406086 DOI: 10.3390/bios12080667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 01/03/2023]
Abstract
Organelles are important subsystems of cells. The damage and inactivation of organelles are closely related to the occurrence of diseases. Organelles’ functional activity can be observed by fluorescence molecular tools. Nowadays, a series of aggregation-induced emission (AIE) bioprobes with organelles-targeting ability have emerged, showing great potential in visualizing the interactions between probes and different organelles. Among them, AIE luminogen (AIEgen)-based peptide bioprobes have attracted more and more attention from researchers due to their good biocompatibility and photostability and abundant diversity. In this review, we summarize the progress of AIEgen-peptide bioprobes in targeting organelles, including the cell membrane, nucleus, mitochondria, lysosomes and endoplasmic reticulum, in recent years. The structural characteristics and biological applications of these bioprobes are discussed, and the development prospect of this field is forecasted. It is hoped that this review will provide guidance for the development of AIEgen-peptide bioprobes at the organelles level and provide a reference for related biomedical research.
Collapse
|
480
|
de Kok NAW, Driessen AJM. The catalytic and structural basis of archaeal glycerophospholipid biosynthesis. Extremophiles 2022; 26:29. [PMID: 35976526 PMCID: PMC9385802 DOI: 10.1007/s00792-022-01277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/02/2022] [Indexed: 12/03/2022]
Abstract
Archaeal glycerophospholipids are the main constituents of the cytoplasmic membrane in the archaeal domain of life and fundamentally differ in chemical composition compared to bacterial phospholipids. They consist of isoprenyl chains ether-bonded to glycerol-1-phosphate. In contrast, bacterial glycerophospholipids are composed of fatty acyl chains ester-bonded to glycerol-3-phosphate. This largely domain-distinguishing feature has been termed the “lipid-divide”. The chemical composition of archaeal membranes contributes to the ability of archaea to survive and thrive in extreme environments. However, ether-bonded glycerophospholipids are not only limited to extremophiles and found also in mesophilic archaea. Resolving the structural basis of glycerophospholipid biosynthesis is a key objective to provide insights in the early evolution of membrane formation and to deepen our understanding of the molecular basis of extremophilicity. Many of the glycerophospholipid enzymes are either integral membrane proteins or membrane-associated, and hence are intrinsically difficult to study structurally. However, in recent years, the crystal structures of several key enzymes have been solved, while unresolved enzymatic steps in the archaeal glycerophospholipid biosynthetic pathway have been clarified providing further insights in the lipid-divide and the evolution of early life.
Collapse
Affiliation(s)
- Niels A W de Kok
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Groningen, The Netherlands
| | - Arnold J M Driessen
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Groningen, The Netherlands.
| |
Collapse
|
481
|
Shrivastava S, Sarkar P, Preira P, Salomé L, Chattopadhyay A. Cholesterol-Dependent Dynamics of the Serotonin 1A Receptor Utilizing Single Particle Tracking: Analysis of Diffusion Modes. J Phys Chem B 2022; 126:6682-6690. [PMID: 35973070 DOI: 10.1021/acs.jpcb.2c03941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) are signaling hubs in cell membranes that regulate a wide range of physiological processes and are popular drug targets. Serotonin1A receptors are important members of the GPCR family and are implicated in neuropsychiatric disorders. Cholesterol is a key constituent of higher eukaryotic membranes and is believed to contribute to the segregated distribution of membrane constituents into domains. To explore the role of cholesterol in lateral dynamics of GPCRs, we utilized single particle tracking (SPT) to monitor diffusion of serotonin1A receptors under acute and chronic cholesterol-depleted conditions. Our results show that the short-term diffusion coefficient of the receptor decreases upon cholesterol depletion, irrespective of the method of cholesterol depletion. Analysis of SPT trajectories revealed that relative populations of receptors undergoing various modes of diffusion change upon cholesterol depletion. Notably, in cholesterol-depleted cells, we observed an increase in the confined population of the receptor accompanied by a reduction in diffusion coefficient for chronic cholesterol depletion. These results are supported by our recent work and present observations that show polymerization of G-actin in response to chronic cholesterol depletion. Taken together, our results bring out the interdependence of cholesterol and actin cytoskeleton in regulating diffusion of GPCRs in membranes.
Collapse
Affiliation(s)
- Sandeep Shrivastava
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Pascal Preira
- Institut de Pharmacologie et de Biologie Structurale, CNRS, Université de Toulouse (UPS), 31 077 Toulouse, France
| | - Laurence Salomé
- Institut de Pharmacologie et de Biologie Structurale, CNRS, Université de Toulouse (UPS), 31 077 Toulouse, France
| | | |
Collapse
|
482
|
HHcy Induces Pyroptosis and Atherosclerosis via the Lipid Raft-Mediated NOX-ROS-NLRP3 Inflammasome Pathway in apoE -/- Mice. Cells 2022; 11:cells11152438. [PMID: 35954287 PMCID: PMC9368640 DOI: 10.3390/cells11152438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/21/2022] Open
Abstract
Lipid rafts play important roles in signal transduction, particularly in responses to inflammatory processes. The current study aimed to identify whether lipid raft-mediated inflammation contributes to hyperhomocysteinemia (HHcy)-accelerated atherosclerosis (AS), and to investigate the underlying mechanisms. THP-1-derived macrophages were used for in vitro experiments. ApoE−/− mice were fed a high-fat diet for 12 weeks to establish an AS model, and a high-fat plus high-methionine diet was used to induce HHcy. We found that homocysteine (Hcy) increased the expression of p22phox and p67phox and promoted their recruitment into lipid rafts (indicating the assembly of the NOX complex), thereby increasing ROS generation and NOX activity, NLRP3 inflammasome activation, and pyroptosis. Mechanistically, Hcy activated the NOX-ROS-NLRP3 inflammasome pathway and induced pyroptosis by increasing the expression of acid sphingomyelinase (ASM) to promote the formation of lipid raft clustering. Importantly, lipid raft-mediated pyroptosis was confirmed in HHcy mice, and HHcy-promoted macrophage recruitment in atherosclerotic lesions and HHcy-aggravated AS were blocked by the lipid raft disruptor methyl-β-cyclodextrin. The study findings indicate that Hcy promotes lipid raft clustering via the upregulation of ASM, which mediates the assembly of the NOX complex, causing an increase in ROS generation, NLRP3 inflammasome activation, and pyroptosis, and contributes to HHcy-induced AS.
Collapse
|
483
|
Chatterjee S, Maltseva D, Kan Y, Hosseini E, Gonella G, Bonn M, Parekh SH. Lipid-driven condensation and interfacial ordering of FUS. SCIENCE ADVANCES 2022; 8:eabm7528. [PMID: 35930639 PMCID: PMC9355348 DOI: 10.1126/sciadv.abm7528] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 06/23/2022] [Indexed: 05/26/2023]
Abstract
Protein condensation into liquid-like structures is critical for cellular compartmentalization, RNA processing, and stress response. Research on protein condensation has primarily focused on membraneless organelles in the absence of lipids. However, the cellular cytoplasm is full of lipid interfaces, yet comparatively little is known about how lipids affect protein condensation. Here, we show that nonspecific interactions between lipids and the disordered fused in sarcoma low-complexity (FUS LC) domain strongly affect protein condensation. In the presence of anionic lipids, FUS LC formed lipid-protein clusters at concentrations more than 30-fold lower than required for pure FUS LC. Lipid-triggered FUS LC clusters showed less dynamic protein organization than canonical, lipid-free FUS LC condensates. Lastly, we found that phosphatidylserine membranes promoted FUS LC condensates having β sheet structures, while phosphatidylglycerol membranes initiated unstructured condensates. Our results show that lipids strongly influence FUS LC condensation, suggesting that protein-lipid interactions modulate condensate formation in cells.
Collapse
Affiliation(s)
- Sayantan Chatterjee
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, TX 78712, USA
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
| | - Daria Maltseva
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
| | - Yelena Kan
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, TX 78712, USA
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
- LUT School of Engineering Science, LUT University, Yliopistonkatu 34, 53850 Lappeenranta, Finland
| | - Elnaz Hosseini
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
| | - Grazia Gonella
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
- Institute of Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Mischa Bonn
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
| | - Sapun H. Parekh
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, TX 78712, USA
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, DE 55128, Germany
| |
Collapse
|
484
|
Jin T, Wu H, Deng Z, Cai T, Li J, Liu Z, Waterhouse PM, White RG, Liang D. Control of root-to-shoot long-distance flow by a key ROS-regulating factor in Arabidopsis. PLANT, CELL & ENVIRONMENT 2022; 45:2476-2491. [PMID: 35689480 DOI: 10.1111/pce.14375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Inter-tissue communication is instrumental to coordinating the whole-body level behaviour for complex multicellular organisms. However, little is known about the regulation of inter-tissue information exchange. Here we carried out genetic screens for root-to-shoot mobile silencing in Arabidopsis plants with a compromised small RNA-mediated gene silencing movement rate and identified radical-induced cell death 1 (RCD1) as a critical regulator of root-shoot communication. RCD1 belongs to a family of poly (ADP-ribose) polymerase proteins, which are highly conserved across land plants. We found that RCD1 coordinates symplastic and apoplastic movement by modulating the sterol level of lipid rafts. The higher superoxide production in rcd1-knockout plants resulted in lower plasmodesmata (PD) frequency and altered PD structure in the symplasm of the hypocotyl cortex. Furthermore, the mutants showed increased lateral area of tracheary pits, which reduced axial movement. Our study highlights a novel mechanism through which root-to-shoot long-distance signalling can be modulated both symplastically and apoplastically.
Collapse
Affiliation(s)
- Tianling Jin
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Huiyan Wu
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Zhuying Deng
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Tingting Cai
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Junkai Li
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Zhangyong Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetlandy, Ministry of Education/Hubei Key Laboratory of Waterlogging Disaster and Wetland Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Peter M Waterhouse
- Centre for Tropical Crops and Biocommodities, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rosemary G White
- Department of Plant Sciences, Australian National University, Canberra, ACT, Australia
| | - Dacheng Liang
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
- Engineering Research Center of Ecology and Agricultural Use of Wetlandy, Ministry of Education/Hubei Key Laboratory of Waterlogging Disaster and Wetland Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
485
|
Lam SM, Huang X, Shui G. Neurological aspects of SARS-CoV-2 infection: lipoproteins and exosomes as Trojan horses. Trends Endocrinol Metab 2022; 33:554-568. [PMID: 35613979 PMCID: PMC9058057 DOI: 10.1016/j.tem.2022.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) primarily targets lipid-producing cells for viral tropism. In this review, we connect systemic lipid couriers, particularly high-density lipoproteins (HDLs) and exosomes, with the neurological facets of SARS-CoV-2 infection. We discuss how SARS-CoV-2 preferentially targets lipid-secreting cells and usurps host cell lipid metabolism for efficient replication and systemic spreading. Besides providing natural veils for viral materials against host immunity, the inherent properties of some of these endogenous lipid particles to traverse the blood-brain barrier (BBB) also offer alternative routes for SARS-CoV-2 neurotropism. Importantly, virus-driven neurological aberrations mediated by HDLs and exosomes are fueled by lipid rafts, which are implicated in the production and transmigration of these lipid particles across the BBB. Finally, we discuss how repurposing existing drugs targeting lipid rafts and cholesterol homeostasis may be beneficial toward alleviating the global coronavirus disease 2019 (COVID-19) disease burden.
Collapse
Affiliation(s)
- Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou 213022, Jiangsu Province, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
486
|
Yano Y. Effects of Membrane Cholesterol on Stability of Transmembrane Helix Associations. Chem Pharm Bull (Tokyo) 2022; 70:514-518. [DOI: 10.1248/cpb.c22-00144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshiaki Yano
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Woman’s University
| |
Collapse
|
487
|
Lunghi G, Carsana EV, Loberto N, Cioccarelli L, Prioni S, Mauri L, Bassi R, Duga S, Straniero L, Asselta R, Soldà G, Di Fonzo A, Frattini E, Magni M, Liessi N, Armirotti A, Ferrari E, Samarani M, Aureli M. β-Glucocerebrosidase Deficiency Activates an Aberrant Lysosome-Plasma Membrane Axis Responsible for the Onset of Neurodegeneration. Cells 2022; 11:cells11152343. [PMID: 35954187 PMCID: PMC9367513 DOI: 10.3390/cells11152343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 02/06/2023] Open
Abstract
β-glucocerebrosidase is a lysosomal hydrolase involved in the catabolism of the sphingolipid glucosylceramide. Biallelic loss of function mutations in this enzyme are responsible for the onset of Gaucher disease, while monoallelic β-glucocerebrosidase mutations represent the first genetic risk factor for Parkinson’s disease. Despite this evidence, the molecular mechanism linking the impairment in β-glucocerebrosidase activity with the onset of neurodegeneration in still unknown. In this frame, we developed two in vitro neuronal models of β-glucocerebrosidase deficiency, represented by mouse cerebellar granule neurons and human-induced pluripotent stem cells-derived dopaminergic neurons treated with the specific β-glucocerebrosidase inhibitor conduritol B epoxide. Neurons deficient for β-glucocerebrosidase activity showed a lysosomal accumulation of glucosylceramide and the onset of neuronal damage. Moreover, we found that neurons react to the lysosomal impairment by the induction of their biogenesis and exocytosis. This latter event was responsible for glucosylceramide accumulation also at the plasma membrane level, with an alteration in lipid and protein composition of specific signaling microdomains. Collectively, our data suggest that β-glucocerebrosidase loss of function impairs the lysosomal compartment, establishing a lysosome–plasma membrane axis responsible for modifications in the plasma membrane architecture and possible alterations of intracellular signaling pathways, leading to neuronal damage.
Collapse
Affiliation(s)
- Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Nicoletta Loberto
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Laura Cioccarelli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Rosaria Bassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (S.D.); (L.S.); (R.A.); (G.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20072 Milan, Italy
| | - Letizia Straniero
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (S.D.); (L.S.); (R.A.); (G.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20072 Milan, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (S.D.); (L.S.); (R.A.); (G.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20072 Milan, Italy
| | - Giulia Soldà
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (S.D.); (L.S.); (R.A.); (G.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20072 Milan, Italy
| | - Alessio Di Fonzo
- IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.D.F.); (E.F.); (M.M.)
| | - Emanuele Frattini
- IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.D.F.); (E.F.); (M.M.)
| | - Manuela Magni
- IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (A.D.F.); (E.F.); (M.M.)
| | - Nara Liessi
- Analytical Chemistry Facility, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (N.L.); (A.A.)
| | - Andrea Armirotti
- Analytical Chemistry Facility, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (N.L.); (A.A.)
| | - Elena Ferrari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy;
| | - Maura Samarani
- Department of Cell Biology and Infection, Institut Pasteur, 75015 Paris, France;
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20054 Milan, Italy; (G.L.); (E.V.C.); (N.L.); (L.C.); (S.P.); (L.M.); (R.B.)
- Correspondence: ; Tel.: +39-025-033-0364
| |
Collapse
|
488
|
Coral Holobionts Possess Distinct Lipid Profiles That May Be Shaped by Symbiodiniaceae Taxonomy. Mar Drugs 2022; 20:md20080485. [PMID: 36005488 PMCID: PMC9410212 DOI: 10.3390/md20080485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Symbiotic relationships are very important for corals. Abiotic stressors cause the acclimatization of cell membranes in symbionts, which possess different membrane acclimatization strategies. Membrane stability is determined by a unique lipid composition and, thus, the profile of thylakoid lipids can depend on coral symbiont species. We have analyzed and compared thylakoid lipidomes (mono- and digalactosyldiacylglycerols (MGDG and DGDG), sulfoquinovosyldiacylglycerols (SQDG), and phosphatidylglycerols (PG)) of crude extracts from symbiotic reef-building coral Acropora sp., the hydrocoral Millepora platyphylla, and the octocoral Sinularia flexibilis. S. flexibilis crude extracts were characterized by a very high SQDG/PG ratio, a DGDG/MGDG ratio < 1, a lower degree of galactolipid unsaturation, a higher content of SQDG with polyunsaturated fatty acids, and a thinner thylakoid membrane which may be explained by the presence of thermosensitive dinoflagellates Cladocopium C3. In contrast, crude extracts of M. platyphylla and Acropora sp. exhibited the lipidome features of thermotolerant Symbiodiniaceae. M. platyphylla and Acropora sp. colonies contained Cladocopium C3u and Cladocopium C71/C71a symbionts, respectively, and their lipidome profiles showed features that indicate thermotolerance. We suggest that an association with symbionts that exhibit the thermotolerant thylakoid lipidome features, combined with a high Symbiodiniaceae diversity, may facilitate further acclimatization/adaptation of M. platyphylla and Acropora sp. holobionts in the South China Sea.
Collapse
|
489
|
Active emulsions in living cell membranes driven by contractile stresses and transbilayer coupling. Proc Natl Acad Sci U S A 2022; 119:e2123056119. [PMID: 35867835 PMCID: PMC9335261 DOI: 10.1073/pnas.2123056119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The spatiotemporal organization of proteins and lipids on the cell surface has direct functional consequences for signaling, sorting, and endocytosis. Earlier studies have shown that multiple types of membrane proteins, including transmembrane proteins that have cytoplasmic actin binding capacity and lipid-tethered glycosylphosphatidylinositol-anchored proteins (GPI-APs), form nanoscale clusters driven by active contractile flows generated by the actin cortex. To gain insight into the role of lipids in organizing membrane domains in living cells, we study the molecular interactions that promote the actively generated nanoclusters of GPI-APs and transmembrane proteins. This motivates a theoretical description, wherein a combination of active contractile stresses and transbilayer coupling drives the creation of active emulsions, mesoscale liquid order (lo) domains of the GPI-APs and lipids, at temperatures greater than equilibrium lipid phase segregation. To test these ideas, we use spatial imaging of molecular clustering combined with local membrane order, and we demonstrate that mesoscopic domains enriched in nanoclusters of GPI-APs are maintained by cortical actin activity and transbilayer interactions and exhibit significant lipid order, consistent with predictions of the active composite model.
Collapse
|
490
|
Piskulich ZA, Cui Q. Machine Learning-Assisted Phase Transition Temperatures from Generalized Replica Exchange Simulations of Dry Martini Lipid Bilayers. J Phys Chem Lett 2022; 13:6481-6486. [PMID: 35819105 DOI: 10.1021/acs.jpclett.2c01654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Accurate estimation of phase transition temperatures has been a longstanding challenge for molecular simulations. Recently, the generalized Replica Exchange technique for estimating phase transition temperatures has allowed for improved sampling of the phase transition; however, it requires a significant number of simultaneous replicas both inside and outside of the transition region leading to costly computational expense. In this work, the recently developed machine learning-assisted lipid phase analysis technique for learning the phase of individual lipids has been combined with generalized Replica Exchange Molecular Dynamics to reduce the overall computational expense of evaluating transition temperatures. This technique is then applied to eight different Dry Martini lipids to demonstrate its ability to describe transition temperatures as a function of chain length and tail saturation.
Collapse
Affiliation(s)
- Zeke A Piskulich
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
491
|
Gupta S, Mandal T. Simulation study of domain formation in a model bacterial membrane. Phys Chem Chem Phys 2022; 24:18133-18143. [PMID: 35856570 DOI: 10.1039/d2cp01873j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent experimental studies revealed that functional membrane microdomains (FMMs) are formed in prokaryotic cells which are structurally and functionally similar to the lipid rafts formed in eukaryotic cells. In this study, we employ coarse-grained molecular dynamics simulations to investigate the mechanism of domain formation and its physiochemical properties in a model methicillin-resistant staphylococcus aureus (MRSA) cell membrane. We find that domains are formed through lateral segregation of staphyloxanthin (STX), a carotenoid which shields the bacteria from the host's immune because of its antioxidant nature. Simulation results suggest that membrane integrity increases with the size of the domain, which is assessed by computing bond order parameter of the lipid tails, membrane expansion modulus and water permeability across the membrane. Various membrane domain proteins such as flotillin-like protein floA and penicillin binding protein (PBP2a) preferentially bind with the STX and accumulate in the membrane domain which is consistent with the recent experimental results.
Collapse
Affiliation(s)
- Shivam Gupta
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur-208016, India.
| | - Taraknath Mandal
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur-208016, India.
| |
Collapse
|
492
|
Lu F, Ferriero DM, Jiang X. Cholesterol in Brain Development and Perinatal Brain Injury: More than a Building Block. Curr Neuropharmacol 2022; 20:1400-1412. [PMID: 34766894 PMCID: PMC9881076 DOI: 10.2174/1570159x19666211111122311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/21/2021] [Accepted: 10/06/2021] [Indexed: 11/22/2022] Open
Abstract
The central nervous system (CNS) is enriched with important classes of lipids, in which cholesterol is known to make up a major portion of myelin sheaths, besides being a structural and functional unit of CNS cell membranes. Unlike in the adult brain, where the cholesterol pool is relatively stable, cholesterol is synthesized and accumulated at the highest rate in the developing brain to meet the needs of rapid brain growth at this stage, which is also a critical period for neuroplasticity. In addition to its biophysical role in membrane organization, cholesterol is crucial for brain development due to its involvement in brain patterning, myelination, neuronal differentiation, and synaptogenesis. Thus any injuries to the immature brain that affect cholesterol homeostasis may have long-term adverse neurological consequences. In this review, we describe the unique features of brain cholesterol biosynthesis and metabolism, cholesterol trafficking between different cell types, and highlight cholesterol-dependent biological processes during brain maturation. We also discuss the association of impaired cholesterol homeostasis with several forms of perinatal brain disorders in term and preterm newborns, including hypoxic-ischemic encephalopathy. Strategies targeting the cholesterol pathways may open new avenues for the diagnosis and treatment of developmental brain injury.
Collapse
Affiliation(s)
- Fuxin Lu
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA;
| | - Donna M. Ferriero
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA; ,Departments of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Xiangning Jiang
- Departments of Neurology, University of California San Francisco, San Francisco, CA, USA; ,Address correspondence to this author at the Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane Room 494, San Francisco, CA 94158, USA; Tel/Fax: 415-502-7285; E-mail:
| |
Collapse
|
493
|
Ge J, Zhang Z, Li Y, Hu Z, He B, Li Y, Zeng B, Jiang C. Inhibition of AoAur1 increases mycelial growth, hyphal fusion and improves physiological adaptation to high-temperature stress in Aspergillus oryzae. Arch Microbiol 2022; 204:477. [PMID: 35829968 DOI: 10.1007/s00203-022-03075-6] [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: 12/23/2021] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 11/02/2022]
Abstract
Inositol phosphorylceramide (IPC) participates in hyphal growth and serves as a signaling molecule that enables fungi to adapt to diverse environments. Here, a gene, encodes IPC synthase, was identified from the Aspergillus oryzae 3.042 genome and designated AoAur1. The characteristics, phylogenetic evolution, and resistance to aureobasidin A of AoAur1 were analyzed. The expression pattern of AoAur1 was markedly downregulated under temperature stress. Additionally, an RNAi-AoAur1 strain in which the AoAur1 expression was inhibited had mycelial that grew more quickly, had a higher frequency of hyphal fusion, and was more resistant to high-temperature stress than the control. Gene expression profiles showed that the genes related to IPC biosynthesis were obviously downregulated, while AoCerS, which participates in dihydroceramide biosynthesis, increased in the RNAi-AoAur1 strain at the three temperature treatments. A metabolomic analysis revealed that the intracellular IPC content decreased, and the accumulation of dihydroceramide and galactosylceramide increased significantly in the RNAi-AoAur1 strain. Thus, the inhibition of AoAur1 reduced IPC level followed by an increase in the contents of dihydroceramide and galactosylceramide that promote mycelial growth and the formation of spores in the RNAi-AoAur1 strain. Interestingly, the inhibition of AoAur1 also induced the expression of hyphal fusion-related genes, which promote hyphal fusion, thus, contributing to the transduction of stress signal to enhance the ability of cells to adapt to temperature stress. Our results demonstrated that the downregulation of AoAur1 and a decrease in the accumulation of IPC is one of the mechanisms that enables A. oryzae to adapt low- and high-temperature stress.
Collapse
Affiliation(s)
- Jinxin Ge
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Zhe Zhang
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yuan Li
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Zhihong Hu
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Bin He
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Yongkai Li
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Bin Zeng
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China. .,College of Pharmacy, Shenzhen Technology University, Shenzhen, 518000, China.
| | - Chunmiao Jiang
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
494
|
Schmieder SS, Tatituri R, Anderson M, Kelly K, Lencer WI. Structural basis for acyl chain control over glycosphingolipid sorting and vesicular trafficking. Cell Rep 2022; 40:111063. [PMID: 35830800 PMCID: PMC9358721 DOI: 10.1016/j.celrep.2022.111063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/13/2021] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
The complex sphingolipids exhibit a diversity of ceramide acyl chain structures that influence their trafficking and intracellular distributions, but it remains unclear how the cell discerns among the different ceramides to affect such sorting. To address the mechanism, we synthesize a library of GM1 glycosphingolipids with naturally varied acyl chains and quantitatively assess their sorting among different endocytic pathways. We find that a stretch of at least 14 saturated carbons extending from C1 at the water-bilayer interface dictate lysosomal sorting by exclusion from endosome sorting tubules. Sorting to the lysosome by the C14∗ motif is cholesterol dependent. Perturbations of the C14∗ motif by unsaturation enable GM1 entry into endosomal sorting tubules of the recycling and retrograde pathways independent of cholesterol. Unsaturation occurring beyond the C14∗ motif in very long acyl chains rescues lysosomal sorting. These results define a structural motif underlying the membrane organization of sphingolipids and implicate cholesterol-sphingolipid nanodomain formation in sorting mechanisms.
Collapse
Affiliation(s)
| | - Raju Tatituri
- Division of Rheumatology, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Michael Anderson
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Digestive Diseases Center, Boston, MA 02115, USA
| | - Kate Kelly
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Wayne I Lencer
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Harvard Digestive Diseases Center, Boston, MA 02115, USA.
| |
Collapse
|
495
|
Zhang S, Zhu N, Gu J, Li HF, Qiu Y, Liao DF, Qin L. Crosstalk between Lipid Rafts and Aging: New Frontiers for Delaying Aging. Aging Dis 2022; 13:1042-1055. [PMID: 35855333 PMCID: PMC9286918 DOI: 10.14336/ad.2022.0116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/16/2022] [Indexed: 12/15/2022] Open
Abstract
With the rapid aging in the global population, delay of aging has become a hot research topic. Lipid rafts (LRs) are microdomains in the plasma membrane that contain sphingolipids and cholesterol. Emerging evidence indicates an interesting interplay between LRs and aging. LRs and their components are altered with aging. Further, the aging process is strongly influenced by LRs. In recent years, LRs and their component signaling molecules have been recognized to affect aging by interfering with its hallmarks. Therefore, targeting LRs is a promising strategy to delay aging.
Collapse
Affiliation(s)
- Shuo Zhang
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Neng Zhu
- 2Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jia Gu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yun Qiu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Li Qin
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.,3Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
496
|
Sviridov D, Miller YI, Bukrinsky MI. Trained Immunity and HIV Infection. Front Immunol 2022; 13:903884. [PMID: 35874772 PMCID: PMC9304701 DOI: 10.3389/fimmu.2022.903884] [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: 03/24/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Findings that certain infections induce immunity not only against the causing agent, but also against an unrelated pathogen have intrigued investigators for many years. Recently, underlying mechanisms of this phenomenon have started to come to light. It was found that the key cells responsible for heterologous protection are innate immune cells such as natural killer cells (NKs), dendritic cells, and monocytes/macrophages. These cells are 'primed' by initial infection, allowing them to provide enhanced response to subsequent infection by the same or unrelated agent. This phenomenon of innate immune memory was termed 'trained immunity'. The proposed mechanism for trained immunity involves activation by the first stimulus of metabolic pathways that lead to epigenetic changes, which maintain the cell in a "trained" state, allowing enhanced responses to a subsequent stimulus. Innate immune memory can lead either to enhanced responses or to suppression of subsequent responses ('tolerance'), depending on the strength and length of the initial stimulation of the immune cells. In the context of HIV infection, innate memory induced by infection is not well understood. In this Hypothesis and Theory article, we discuss evidence for HIV-induced trained immunity in human monocytes, its possible mechanisms, and implications for HIV-associated co-morbidities.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Laboratory of Lipoproteins and Atherosclerosis, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Michael I. Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
497
|
Skryabin GO, Vinokurova SV, Galetsky SA, Elkin DS, Senkovenko AM, Denisova DA, Komelkov AV, Stilidi IS, Peregorodiev IN, Malikhova OA, Imaraliev OT, Enikeev AD, Tchevkina EM. Isolation and Characterization of Extracellular Vesicles from Gastric Juice. Cancers (Basel) 2022; 14:cancers14143314. [PMID: 35884376 PMCID: PMC9318556 DOI: 10.3390/cancers14143314] [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: 05/27/2022] [Revised: 06/24/2022] [Accepted: 07/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Gastric cancer (GC) is one of the most common cancers and the fifth leading cause of cancer-related deaths worldwide. The steadily growing interest in secreted extracellular vesicles (EVs) is related to their ability to carry a variety of biologically active molecules, which can be used as markers for liquid noninvasive diagnosis of malignant neoplasms. For these applications, blood is the most widely used source of EVs. However, this body fluid contains an extremely heterogeneous mixture of EVs originating from different types of normal cells and tissues. The aim of this study was to assess the possibility of using gastric juice (GJ) as an alternative source of EVs since it is expected to be enriched in vesicles of tumor origin. We validated the presence of EVs in GJ using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western-blot analysis of exosomal markers, showed for the first time the feasibility of their isolation by ultracentrifugation and demonstrated the prospect of using GJ-derived EVs as a source of GC miRNA markers. Abstract EVs are involved in local and distant intercellular communication and play a vital role in cancer development. Since EVs have been found in almost all body fluids, there are currently active attempts for their application in liquid diagnostics. Blood is the most commonly used source of EVs for the screening of cancer markers, although the percentage of tumor-derived EVs in the blood is extremely low. In contrast, GJ, as a local biofluid, is expected to be enriched with GC-associated EVs. However, EVs from GJ have never been applied for the screening and are underinvestigated overall. Here we show that EVs can be isolated from GJ by ultracentrifugation. TEM analysis showed high heterogeneity of GJ-derived EVs, including those with exosome-like size and morphology. In addition to morphological diversity, EVs from individual GJ samples differed in the composition of exosomal markers. We also show the presence of stomatin within GJ-derived EVs for the first time. The first conducted comparison of miRNA content in EVs from GC patients and healthy donors performed using a pilot sampling revealed the significant differences in several miRNAs (-135b-3p, -199a-3p, -451a). These results demonstrate the feasibility of the application of GJ-derived EVs for screening for miRNA GC markers.
Collapse
Affiliation(s)
- Gleb O. Skryabin
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Svetlana V. Vinokurova
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Sergey A. Galetsky
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Danila S. Elkin
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Alexey M. Senkovenko
- Department of Bioengineering, Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory, 1/12, 111234 Moscow, Russia;
| | - Darya A. Denisova
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Andrey V. Komelkov
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
- Correspondence: (A.V.K.); (E.M.T.)
| | - Ivan S. Stilidi
- Research Institute of Clinical Oncology, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (I.S.S.); (I.N.P.); (O.A.M.); (O.T.I.)
| | - Ivan N. Peregorodiev
- Research Institute of Clinical Oncology, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (I.S.S.); (I.N.P.); (O.A.M.); (O.T.I.)
| | - Olga A. Malikhova
- Research Institute of Clinical Oncology, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (I.S.S.); (I.N.P.); (O.A.M.); (O.T.I.)
| | - Oiatiddin T. Imaraliev
- Research Institute of Clinical Oncology, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (I.S.S.); (I.N.P.); (O.A.M.); (O.T.I.)
| | - Adel D. Enikeev
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
| | - Elena M. Tchevkina
- Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoye Sh. 24, 115478 Moscow, Russia; (G.O.S.); (S.V.V.); (S.A.G.); (D.S.E.); (D.A.D.); (A.D.E.)
- Correspondence: (A.V.K.); (E.M.T.)
| |
Collapse
|
498
|
Contribution of specific ceramides to obesity-associated metabolic diseases. Cell Mol Life Sci 2022; 79:395. [PMID: 35789435 PMCID: PMC9252958 DOI: 10.1007/s00018-022-04401-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022]
Abstract
Ceramides are a heterogeneous group of bioactive membrane sphingolipids that play specialized regulatory roles in cellular metabolism depending on their characteristic fatty acyl chain lengths and subcellular distribution. As obesity progresses, certain ceramide molecular species accumulate in metabolic tissues and cause cell-type-specific lipotoxic reactions that disrupt metabolic homeostasis and lead to the development of cardiometabolic diseases. Several mechanisms for ceramide action have been inferred from studies in vitro, but only recently have we begun to better understand the acyl chain length specificity of ceramide-mediated signaling in the context of physiology and disease in vivo. New discoveries show that specific ceramides affect various metabolic pathways and that global or tissue-specific reduction in selected ceramide pools in obese rodents is sufficient to improve metabolic health. Here, we review the tissue-specific regulation and functions of ceramides in obesity, thus highlighting the emerging concept of selectively inhibiting production or action of ceramides with specific acyl chain lengths as novel therapeutic strategies to ameliorate obesity-associated diseases.
Collapse
|
499
|
Tourte M, Coffinet S, Wörmer L, Lipp JS, Hinrichs KU, Oger PM. The Exploration of the Thermococcus barophilus Lipidome Reveals the Widest Variety of Phosphoglycolipids in Thermococcales. Front Microbiol 2022; 13:869479. [PMID: 35865931 PMCID: PMC9294538 DOI: 10.3389/fmicb.2022.869479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
One of the most distinctive characteristics of archaea is their unique lipids. While the general nature of archaeal lipids has been linked to their tolerance to extreme conditions, little is known about the diversity of lipidic structures archaea are able to synthesize, which hinders the elucidation of the physicochemical properties of their cell membrane. In an effort to widen the known lipid repertoire of the piezophilic and hyperthermophilic model archaeon Thermococcus barophilus, we comprehensively characterized its intact polar lipid (IPL), core lipid (CL), and polar head group compositions using a combination of cutting-edge liquid chromatography and mass spectrometric ionization systems. We tentatively identified 82 different IPLs based on five distinct CLs and 10 polar head group derivatives of phosphatidylhexoses, including compounds reported here for the first time, e.g., di-N-acetylhexosamine phosphatidylhexose-bearing lipids. Despite having extended the knowledge on the lipidome, our results also indicate that the majority of T. barophilus lipids remain inaccessible to current analytical procedures and that improvements in lipid extraction and analysis are still required. This expanded yet incomplete lipidome nonetheless opens new avenues for understanding the physiology, physicochemical properties, and organization of the membrane in this archaeon as well as other archaea.
Collapse
Affiliation(s)
- Maxime Tourte
- Univ. Lyon, Univ. Lyon 1, CNRS, UMR 5240, Villeurbanne, France
- Univ. Lyon, INSA Lyon, CNRS, UMR 5240, Villeurbanne, France
| | - Sarah Coffinet
- MARUM Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | - Lars Wörmer
- MARUM Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | - Julius S. Lipp
- MARUM Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | - Kai-Uwe Hinrichs
- MARUM Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | | |
Collapse
|
500
|
Taoro-González L, Pereda D, Valdés-Baizabal C, González-Gómez M, Pérez JA, Mesa-Herrera F, Canerina-Amaro A, Pérez-González H, Rodríguez C, Díaz M, Marin R. Effects of Dietary n-3 LCPUFA Supplementation on the Hippocampus of Aging Female Mice: Impact on Memory, Lipid Raft-Associated Glutamatergic Receptors and Neuroinflammation. Int J Mol Sci 2022; 23:7430. [PMID: 35806435 PMCID: PMC9267073 DOI: 10.3390/ijms23137430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 11/17/2022] Open
Abstract
Long-chain polyunsaturated fatty acids (LCPUFA), essential molecules whose precursors must be dietary supplied, are highly represented in the brain contributing to numerous neuronal processes. Recent findings have demonstrated that LCPUFA are represented in lipid raft microstructures, where they favor molecular interactions of signaling complexes underlying neuronal functionality. During aging, the brain lipid composition changes affecting the lipid rafts' integrity and protein signaling, which may induce memory detriment. We investigated the effect of a n-3 LCPUFA-enriched diet on the cognitive function of 6- and 15-months-old female mice. Likewise, we explored the impact of dietary n-3 LCPUFAs on hippocampal lipid rafts, and their potential correlation with aging-induced neuroinflammation. Our results demonstrate that n-3 LCPUFA supplementation improves spatial and recognition memory and restores the expression of glutamate and estrogen receptors in the hippocampal lipid rafts of aged mice to similar profiles than young ones. Additionally, the n-3 LCPUFA-enriched diet stabilized the lipid composition of the old mice's hippocampal lipid rafts to the levels of young ones and reduced the aged-induced neuroinflammatory markers. Hence, we propose that n-3 LCPUFA supplementation leads to beneficial cognitive performance by "rejuvenating" the lipid raft microenvironment that stabilizes the integrity and interactions of memory protein players embedded in these microdomains.
Collapse
Affiliation(s)
- Lucas Taoro-González
- Research Unit, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain;
- Instituto de Tecnologías Biomédicas de Canarias (ITB), University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (M.G.-G.); (C.R.)
| | - Daniel Pereda
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (D.P.); (C.V.-B.); (A.C.-A.)
- Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Catalina Valdés-Baizabal
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (D.P.); (C.V.-B.); (A.C.-A.)
- Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Miriam González-Gómez
- Instituto de Tecnologías Biomédicas de Canarias (ITB), University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (M.G.-G.); (C.R.)
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
- Instituto de Neurociencia Cognitiva (NeuroCog), University of La Laguna, 38205 San Cristóbal de La Laguna, Spain
| | - José A. Pérez
- Department of Animal Biology, Edaphology and Geology, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
| | - Fátima Mesa-Herrera
- Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
| | - Ana Canerina-Amaro
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (D.P.); (C.V.-B.); (A.C.-A.)
- Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Herminia Pérez-González
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
| | - Covadonga Rodríguez
- Instituto de Tecnologías Biomédicas de Canarias (ITB), University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (M.G.-G.); (C.R.)
- Department of Animal Biology, Edaphology and Geology, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
| | - Mario Díaz
- Instituto de Neurociencia Cognitiva (NeuroCog), University of La Laguna, 38205 San Cristóbal de La Laguna, Spain
- Department of Physics, Faculty of Sciences, University of La Laguna, 38200 San Cristóbal de La Laguna, Spain
- IUETSP (Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias), University of La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Section of Medicine, Faculty of Health Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain; (D.P.); (C.V.-B.); (A.C.-A.)
- Associate Research Unit ULL-CSIC, Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain
| |
Collapse
|