1
|
Karbanová J, Thamm K, Fargeas CA, Deniz IA, Lorico A, Corbeil D. Prominosomes - a particular class of extracellular vesicles containing prominin-1/CD133? J Nanobiotechnology 2025; 23:61. [PMID: 39881297 PMCID: PMC11776279 DOI: 10.1186/s12951-025-03102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Extracellular membrane vesicles (EVs) offer promising values in various medical fields, e.g., as biomarkers in liquid biopsies or as native (or bioengineered) biological nanocarriers in tissue engineering, regenerative medicine and cancer therapy. Based on their cellular origin EVs can vary considerably in composition and diameter. Cell biological studies on mammalian prominin-1, a cholesterol-binding membrane glycoprotein, have helped to reveal new donor membranes as sources of EVs. For instance, small EVs can originate from microvilli and primary cilia, while large EVs might be produced by transient structures such as retracting cellular extremities of cancer cells during the mitotic rounding process, and the midbody at the end of cytokinesis. Here, we will highlight the various subcellular origins of prominin-1+ EVs, also called prominosomes, and the potential mechanism(s) regulating their formation. We will further discuss the molecular and cellular characteristics of prominin-1, notably those that have a direct effect on the release of prominin-1+ EVs, a process that might be directly implicated in donor cell reprogramming of stem and cancer stem cells. Prominin-1+ EVs also mediate intercellular communication during embryonic development and adult homeostasis in healthy individuals, while disseminating biological information during diseases.
Collapse
Affiliation(s)
- Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| | - Kristina Thamm
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- denovoMATRIX GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Christine A Fargeas
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ilker A Deniz
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV, 89014, USA
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| |
Collapse
|
2
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Maggiorani D, Santin Y, Formoso K, Drapé E, Martini H, Brun S, Cousin G, Lairez O, Lezoualc'h F, Parini A, Douin‐Echinard V, Mialet‐Perez J. Identification of Prominin-2 as a new player of cardiomyocyte senescence in the aging heart. Aging Cell 2024; 23:e14204. [PMID: 38757782 PMCID: PMC11488343 DOI: 10.1111/acel.14204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
The aging heart is characterized by a number of structural changes leading to ventricular stiffness, impaired resistance to stress and increased risk of developing heart failure (HF). Genetic or pharmacological removal of senescent cells has recently demonstrated the possibility to relieve some cardiac aging features such as hypertrophy and fibrosis. However, the contribution of the different cell types in cardiac aging remains fragmentary due to a lack of cell-specific markers. Cardiomyocytes undergo post-mitotic senescence in response to telomere damage, characterized by persistent DNA damage response and expression of the classical senescence markers p21 and p16, which are shared by many other cell types. In the present study, we used transcriptomic approaches to discover new markers specific for cardiomyocyte senescence. We identified Prominin2 (Prom2), encoding a transmembrane glycoprotein, as the most upregulated gene in cardiomyocytes of aged mice compared to young mice. We showed that Prom2 was upregulated by a p53-dependent pathway in stress-induced premature senescence. Prom2 expression correlated with cardiomyocyte hypertrophy in the hearts of aged mice and was increased in atrial samples of patients with HF with preserved ejection fraction. Consistently, Prom2 overexpression was sufficient to drive senescence, hypertrophy and resistance to cytotoxic stress while Prom2 shRNA silencing inhibited these features in doxorubicin-treated cardiac cells. In conclusion, we identified Prom2 as a new player of cardiac aging, linking cardiomyocyte hypertrophy to senescence. These results could provide a better understanding and targeting of cell-type specific senescence in age-associated cardiac diseases.
Collapse
Affiliation(s)
- D. Maggiorani
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - Y. Santin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - K. Formoso
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - E. Drapé
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - H. Martini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- Rangueil Hospital, CHUToulouseFrance
| | - S. Brun
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- Rangueil Hospital, CHUToulouseFrance
| | - G. Cousin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- Rangueil Hospital, CHUToulouseFrance
| | - O. Lairez
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- Rangueil Hospital, CHUToulouseFrance
| | - F. Lezoualc'h
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - A. Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
| | - V. Douin‐Echinard
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- RESTORE Research Center, UMR‐1301, INSERM, CNRS, EFSUniversity of ToulouseToulouseFrance
| | - J. Mialet‐Perez
- Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR‐1297 INSERM, University of ToulouseToulouseFrance
- Univ Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| |
Collapse
|
4
|
Paris J, Wilhelm C, Lebbé C, Elmallah M, Pamoukdjian F, Héraud A, Gapihan G, Walle AVD, Tran VN, Hamdan D, Allayous C, Battistella M, Van Glabeke E, Lim KW, Leboeuf C, Roger S, Falgarone G, Phan AT, Bousquet G. PROM2 overexpression induces metastatic potential through epithelial-to-mesenchymal transition and ferroptosis resistance in human cancers. Clin Transl Med 2024; 14:e1632. [PMID: 38515278 PMCID: PMC10958126 DOI: 10.1002/ctm2.1632] [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] [Received: 10/13/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Despite considerable therapeutic advances in the last 20 years, metastatic cancers remain a major cause of death. We previously identified prominin-2 (PROM2) as a biomarker predictive of distant metastases and decreased survival, thus providing a promising bio-target. In this translational study, we set out to decipher the biological roles of PROM2 during the metastatic process and resistance to cell death, in particular for metastatic melanoma. METHODS AND RESULTS Methods and results: We demonstrated that PROM2 overexpression was closely linked to an increased metastatic potential through the increase of epithelial-to-mesenchymal transition (EMT) marker expression and ferroptosis resistance. This was also found in renal cell carcinoma and triple negative breast cancer patient-derived xenograft models. Using an oligonucleotide anti-sense anti-PROM2, we efficaciously decreased PROM2 expression and prevented metastases in melanoma xenografts. We also demonstrated that PROM2 was implicated in an aggravation loop, contributing to increase the metastatic burden both in murine metastatic models and in patients with metastatic melanoma. The metastatic burden is closely linked to PROM2 expression through the expression of EMT markers and ferroptosis cell death resistance in a deterioration loop. CONCLUSION Our results open the way for further studies using PROM2 as a bio-target in resort situations in human metastatic melanoma and also in other cancer types.
Collapse
Affiliation(s)
- Justine Paris
- Université Paris Cité, INSERM, UMR_S942 MASCOTParisFrance
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie, Institut Curie, CNRSPSL Research UniversityParisFrance
| | - Celeste Lebbé
- Université Paris Cité, INSERMParisFrance
- APHP, Dermatolo‐OncologyHôpital Saint LouisParisFrance
| | - Mohammed Elmallah
- Inserm U1327 ISCHEMIAUniversité de Tours, Faculté de MédecineToursFrance
| | - Frédéric Pamoukdjian
- Université Paris Cité, INSERM, UMR_S942 MASCOTParisFrance
- APHP, Hôpital Avicenne, Médecine GériatriqueBobignyFrance
- Université Sorbonne Paris NordVilletaneuseFrance
| | - Audrey Héraud
- Inserm U1327 ISCHEMIAUniversité de Tours, Faculté de MédecineToursFrance
| | | | - Aurore Van De Walle
- Laboratoire Physico Chimie Curie, Institut Curie, CNRSPSL Research UniversityParisFrance
| | - Van Nhan Tran
- School of Physical and Mathematical SciencesNanyang Technological UniversitySingaporeSingapore
| | - Diaddin Hamdan
- Université Paris Cité, INSERM, UMR_S942 MASCOTParisFrance
- Hôpital La Porte Verte, CancérologieVersaillesFrance
| | - Clara Allayous
- Université Paris Cité, INSERMParisFrance
- APHP, Dermatolo‐OncologyHôpital Saint LouisParisFrance
| | - Maxime Battistella
- Université Paris Cité, INSERMParisFrance
- Pathology DepartmentAPHP, Hôpital Saint LouisParisFrance
| | - Emmanuel Van Glabeke
- Fédération d'Urologie de Seine‐Saint‐Denis, CHI Robert BallangéAulnay‐sous‐BoisFrance
| | - Kah Wai Lim
- School of Physical and Mathematical SciencesNanyang Technological UniversitySingaporeSingapore
| | | | - Sébastien Roger
- Inserm U1327 ISCHEMIAUniversité de Tours, Faculté de MédecineToursFrance
| | - Géraldine Falgarone
- Université Paris Cité, INSERM, UMR_S942 MASCOTParisFrance
- APHP, Hôpital Avicenne, Médecine GériatriqueBobignyFrance
- APHPHôpital Avicenne, Unité de Médecine Ambulatoire (UMA)BobignyFrance
| | - Anh Tuan Phan
- Université Sorbonne Paris NordVilletaneuseFrance
- NTU Institute of Structural BiologyNanyang Technological UniversitySingaporeSingapore
| | - Guilhem Bousquet
- Université Paris Cité, INSERM, UMR_S942 MASCOTParisFrance
- APHP, Hôpital Avicenne, Médecine GériatriqueBobignyFrance
- APHPHôpital Avicenne, Oncologie médicalBobignyFrance
| |
Collapse
|
5
|
Activation of Non-Canonical Autophagic Pathway through Inhibition of Non-Integrin Laminin Receptor in Neuronal Cells. Cells 2022; 11:cells11030466. [PMID: 35159276 PMCID: PMC8833926 DOI: 10.3390/cells11030466] [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: 10/06/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/05/2022] Open
Abstract
To fight neurodegenerative diseases, several therapeutic strategies have been proposed that, to date, are either ineffective or at the early preclinical stages. Intracellular protein aggregates represent the cause of about 70% of neurodegenerative disorders, such as Alzheimer’s disease. Thus, autophagy, i.e., lysosomal degradation of macromolecules, could be employed in this context as a therapeutic strategy. Searching for a compound that stimulates this process led us to the identification of a 37/67kDa laminin receptor inhibitor, NSC48478. We have analysed the effects of this small molecule on the autophagic process in mouse neuronal cells and found that NSC48478 induces the conversion of microtubule-associated protein 1A/1B-light chain 3 (LC3-I) into the LC3-phosphatidylethanolamine conjugate (LC3-II). Interestingly, upon NSC48478 treatment, the contribution of membranes to the autophagic process derived mainly from the non-canonical m-TOR-independent endocytic pathway, involving the Rab proteins that control endocytosis and vesicle recycling. Finally, qRT-PCR analysis suggests that, while the expression of key genes linked to canonical autophagy was unchanged, the main genes related to the positive regulation of endocytosis (pinocytosis and receptor mediated), along with genes regulating vesicle fusion and autolysosomal maturation, were upregulated under NSC48478 conditions. These results strongly suggest that 37/67 kDa inhibitor could be a useful tool for future studies in pathological conditions.
Collapse
|
6
|
Jozic I, Abujamra BA, Elliott MH, Wikramanayake TC, Marjanovic J, Stone RC, Head CR, Pastar I, Kirsner RS, Andreopoulos FM, Musi JP, Tomic-Canic M. Glucocorticoid-mediated induction of caveolin-1 disrupts cytoskeletal organization, inhibits cell migration and re-epithelialization of non-healing wounds. Commun Biol 2021; 4:757. [PMID: 34145387 PMCID: PMC8213848 DOI: 10.1038/s42003-021-02298-5] [Citation(s) in RCA: 16] [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: 03/11/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
Although impaired keratinocyte migration is a recognized hallmark of chronic wounds, the molecular mechanisms underpinning impaired cell movement are poorly understood. Here, we demonstrate that both diabetic foot ulcers (DFUs) and venous leg ulcers (VLUs) exhibit global deregulation of cytoskeletal organization in genomic comparison to normal skin and acute wounds. Interestingly, we found that DFUs and VLUs exhibited downregulation of ArhGAP35, which serves both as an inactivator of RhoA and as a glucocorticoid repressor. Since chronic wounds exhibit elevated levels of cortisol and caveolin-1 (Cav1), we posited that observed elevation of Cav1 expression may contribute to impaired actin-cytoskeletal signaling, manifesting in aberrant keratinocyte migration. We showed that Cav1 indeed antagonizes ArhGAP35, resulting in increased activation of RhoA and diminished activation of Cdc42, which can be rescued by Cav1 disruption. Furthermore, we demonstrate that both inducible keratinocyte specific Cav1 knockout mice, and MβCD treated diabetic mice, exhibit accelerated wound closure. Taken together, our findings provide a previously unreported mechanism by which Cav1-mediated cytoskeletal organization prevents wound closure in patients with chronic wounds.
Collapse
Affiliation(s)
- Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Beatriz Abdo Abujamra
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael H Elliott
- Departments of Ophthalmology, Physiology, and Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tongyu C Wikramanayake
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Cheyanne R Head
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert S Kirsner
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fotios M Andreopoulos
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan P Musi
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
7
|
Sepers B, Erven JAM, Gawehns F, Laine VN, van Oers K. Epigenetics and Early Life Stress: Experimental Brood Size Affects DNA Methylation in Great Tits (Parus major). Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.609061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Early developmental conditions are known to have life-long effects on an individual’s behavior, physiology and fitness. In altricial birds, a majority of these conditions, such as the number of siblings and the amount of food provisioned, are controlled by the parents. This opens up the potential for parents to adjust the behavior and physiology of their offspring according to local post-natal circumstances. However, the mechanisms underlying such intergenerational regulation remain largely unknown. A mechanism often proposed to possibly explain how parental effects mediate consistent phenotypic change is DNA methylation. To investigate whether early life effects on offspring phenotypes are mediated by DNA methylation, we cross-fostered great tit (Parus major) nestlings and manipulated their brood size in a natural study population. We assessed genome-wide DNA methylation levels of CpG sites in erythrocyte DNA, using Reduced Representation Bisulfite Sequencing (RRBS). By comparing DNA methylation levels between biological siblings raised in enlarged and reduced broods and between biological siblings of control broods, we assessed which CpG sites were differentially methylated due to brood size. We found 32 differentially methylated sites (DMS) between siblings from enlarged and reduced broods, a larger number than in the comparison between siblings from control broods. A considerable number of these DMS were located in or near genes involved in development, growth, metabolism, behavior and cognition. Since the biological functions of these genes line up with previously found effects of brood size and food availability, it is likely that the nestlings in the enlarged broods suffered from nutritional stress. We therefore conclude that early life stress might directly affect epigenetic regulation of genes related to early life conditions. Future studies should link such experimentally induced DNA methylation changes to expression of phenotypic traits and assess whether these effects affect parental fitness to determine if such changes are also adaptive.
Collapse
|
8
|
Nguyen TT, Gapihan G, Tetu P, Pamoukdjian F, El Bouchtaoui M, Lebœuf C, Feugeas JP, Paris J, Baroudjian B, Delyon J, Mourah S, Lebbé C, Janin A, Bousquet G, Battistella M. Increased risk of brain metastases among patients with melanoma and PROM2 expression in metastatic lymph nodes. Clin Transl Med 2020; 10:e198. [PMID: 33377645 PMCID: PMC7711084 DOI: 10.1002/ctm2.198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 11/28/2022] Open
Affiliation(s)
- Thuy Thi Nguyen
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France.,AP-HP Hôpital Avicenne, Oncologie Médicale, Bobigny, France.,Medical Oncology Department A, National Cancer Hospital, Hanoi, Vietnam
| | - Guillaume Gapihan
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France.,Hôpital Saint-Louis, Université de Paris, Paris, France
| | - Pauline Tetu
- Dermatologie, AP-HP Hôpital Saint Louis, Paris, France
| | - Frédéric Pamoukdjian
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France.,Médecine Gériatrique, AP-HP Hôpital Avicenne, Bobigny, France.,Université Sorbonne Paris Nord, Cardiovascular Markers in Stressed Conditions, MASCOT, Bobigny, France
| | | | - Christophe Lebœuf
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France
| | - Jean-Paul Feugeas
- INSERM U722, Paris, France.,Université de Franche-Comté, Besançon, France
| | - Justine Paris
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France
| | | | - Julie Delyon
- Dermatologie, AP-HP Hôpital Saint Louis, Paris, France.,INSERM U976, Paris, France
| | - Samia Mourah
- Hôpital Saint-Louis, Université de Paris, Paris, France.,INSERM U976, Paris, France.,Solid Tumor Genomic Department, AP-HP, Hôpital Saint Louis, Paris, France
| | - Céleste Lebbé
- Hôpital Saint-Louis, Université de Paris, Paris, France.,Dermatologie, AP-HP Hôpital Saint Louis, Paris, France.,INSERM U976, Paris, France
| | - Anne Janin
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France.,Hôpital Saint-Louis, Université de Paris, Paris, France
| | - Guilhem Bousquet
- INSERM U942, Universités de Paris et Sorbonne Paris Nord, Bobigny, France.,AP-HP Hôpital Avicenne, Oncologie Médicale, Bobigny, France.,Université Sorbonne Paris Nord, Cardiovascular Markers in Stressed Conditions, MASCOT, Bobigny, France
| | - Maxime Battistella
- Hôpital Saint-Louis, Université de Paris, Paris, France.,INSERM U976, Paris, France.,Laboratoire de Pathologie, AP-HP Hôpital Saint-Louis, Paris, France
| |
Collapse
|
9
|
Saha SK, Islam SMR, Kwak KS, Rahman MS, Cho SG. PROM1 and PROM2 expression differentially modulates clinical prognosis of cancer: a multiomics analysis. Cancer Gene Ther 2020; 27:147-167. [PMID: 31164716 PMCID: PMC7170805 DOI: 10.1038/s41417-019-0109-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/03/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Prominin 1 (PROM1) is considered a biomarker for cancer stem cells, although its biological role is unclear. Prominin 2 (PROM2) has also been associated with certain cancers. However, the prognostic value of PROM1 and PROM2 in cancer is controversial. Here, we performed a systematic data analysis to examine whether prominins can function as prognostic markers in human cancers. The expression of prominins was assessed and their prognostic value in human cancers was determined using univariate and multivariate survival analyses, via various online platforms. We selected a group of prominent functional protein partners of prominins by protein-protein interaction analysis. Subsequently, we investigated the relationship between mutations and copy number alterations in prominin genes and various types of cancers. Furthermore, we identified genes that correlated with PROM1 and PROM2 in certain cancers, based on their levels of expression. Gene ontology and pathway analyses were performed to assess the effect of these correlated genes on various cancers. We observed that PROM1 was frequently overexpressed in esophageal, liver, and ovarian cancers and its expression was negatively associated with prognosis, whereas PROM2 overexpression was associated with poor overall survival in lung and ovarian cancers. Based on the varying characteristics of prominins, we conclude that PROM1 and PROM2 expression differentially modulates the clinical outcomes of cancers.
Collapse
Affiliation(s)
- Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| | - S M Riazul Islam
- Department of Computer Science and Engineering, Sejong University, 209, Neungdong-ro, Gwangjin-gu, Seoul, 05006, Republic of Korea
| | - Kyung-Sup Kwak
- School of Information and Communication Engineering, Inha University, 100, Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea
| | - Md Shahedur Rahman
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
10
|
Li W, Zhu Y, Zhang K, Yu X, Lin H, Wu W, Peng Y, Sun J. PROM2 promotes gemcitabine chemoresistance via activating the Akt signaling pathway in pancreatic cancer. Exp Mol Med 2020; 52:409-422. [PMID: 32123287 PMCID: PMC7156657 DOI: 10.1038/s12276-020-0390-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/08/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
In recent years, the deoxycytidine analogue gemcitabine (2′,2′,-difluorodeoxycytidine) has become the first-line chemotherapeutic agent for patients with pancreatic cancer. However, due to the intrinsic resistance of pancreatic cancer cells, gemcitabine-based chemotherapy yields limited disease control, with >85% disease progression at 6 months from diagnosis. Therefore, elucidating the mechanisms of chemoresistance is a critical step in improving cancer therapy, especially for the treatment of pancreatic cancer. We show PROM2, a transmembrane glycoprotein, is ubiquitously upregulated in pancreatic cancer cell. We also found higher PROM2 expression is associated with shortened overall and disease-free survival times in patients diagnosed with pancreatic cancer. We provide evidence that PROM2 promotes chemoresistance to gemcitabine both in vivo and in vitro. Mechanistically, we demonstrate that PROM2 could directly interacted with Akt and activates the Akt signaling pathway, which thus inhibiting gemcitabine-induced apoptosis. As further evidence, we show PROM2 expression and Akt phosphorylation both promote gemcitabine chemoresistance, and cause poorer survival in clinical samples with pancreatic cancer. Combining gemcitabine with the Akt inhibitor MK-2206 facilitated significant tumor shrinkage and dramatically elevated the survival status in mice xenografted with pancreatic cancer cells. Our findings not only establish PROM2 as a novel positive regulator of the Akt signaling pathway and a candidate prognostic indicator of gemcitabine response, but also provide a neo-therapeutic approach for patients resistant to gemcitabine treatment. A cell membrane protein called PROM2 promotes the resistance of pancreatic cancer to the anti-cancer drug gemcitabine, suggesting PROM2 and the molecular signaling pathway it stimulates could be targeted by new treatments. Researchers in China led by Jian Sun at Sun Yat-Sen University, Guangzhou, investigated the role of PROM2 in cultured human pancreatic cancer cells and in a mouse model of pancreatic cancer. Production and activity of PROM2 were increased in cancer cells, leading to increased resistance to gemcitabine. The researchers found that PROM2’s promotion of gemcitabine resistance was linked to its ability to bind to another protein called Akt. This interaction stimulates the Akt signaling pathway, sustaining cancer cells. Combining gemcitabine therapy with an Akt pathway inhibitor restored cancer cell sensitivity to gemcitabine, revealing a potential approach to developing drugs to overcome gemcitabine resistance.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yue Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Department of Vascular and Thyroid Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kelin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Department of Surgical Intensive Care Unit, Sun Yat‑sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Xianhuan Yu
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Haoming Lin
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wenrui Wu
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yaorong Peng
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Jian Sun
- Department of Hepatobiliary and pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
11
|
Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, Baer CE, Dixon SJ, Mercurio AM. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev Cell 2019; 51:575-586.e4. [PMID: 31735663 PMCID: PMC8316835 DOI: 10.1016/j.devcel.2019.10.007] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/28/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022]
Abstract
Ferroptosis, regulated cell death characterized by the iron-dependent accumulation of lethal lipid reactive oxygen species, contributes to tissue homeostasis and numerous pathologies, and it may be exploited for therapy. Cells differ in their sensitivity to ferroptosis, however, and a key challenge is to understand mechanisms that contribute to resistance. Using RNA-seq to identify genes that contribute to ferroptosis resistance, we discovered that pro-ferroptotic stimuli, including inhibition of the lipid hydroperoxidase GPX4 and detachment from the extracellular matrix, induce expression of prominin2, a pentaspanin protein implicated in regulation of lipid dynamics. Prominin2 facilitates ferroptosis resistance in mammary epithelial and breast carcinoma cells. Mechanistically, prominin2 promotes the formation of ferritin-containing multivesicular bodies (MVBs) and exosomes that transport iron out of the cell, inhibiting ferroptosis. These findings reveal that ferroptosis resistance can be driven by a prominin2-MVB-exosome-ferritin pathway and have broad implications for iron homeostasis, intracellular trafficking, and cancer.
Collapse
Affiliation(s)
- Caitlin W Brown
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - John J Amante
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Peter Chhoy
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ameer L Elaimy
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Haibo Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Christina E Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA; Sanderson Center for Optical Examination, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
12
|
Dowland SN, Madawala RJ, Poon CE, Lindsay LA, Murphy CR. Prominin-2 Prevents the Formation of Caveolae in Normal and Ovarian Hyperstimulated Pregnancy. Reprod Sci 2017; 25:1231-1242. [PMID: 29113580 DOI: 10.1177/1933719117737842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
During early pregnancy, uterine epithelial cells (UECs) become less adherent to the underlying basal lamina and are subsequently removed so the blastocyst can invade the underlying stroma. This process involves the removal of focal adhesions from the basal plasma membrane of UECs. These focal adhesions are thought to be internalized by caveolae, which significantly increase in abundance at the time of blastocyst implantation. A recent in vitro study indicated that prominin-2 prevents the formation of caveolae by sequestering membrane cholesterol. The present study examines whether prominin-2 affects the formation of caveolae and loss of focal adhesions in UECs during normal and ovarian hyperstimulation (OH) pregnancy in the rat. At the time of fertilization during normal pregnancy, prominin-2 is distributed throughout the basolateral plasma membrane. However, at the time of implantation and coincident with an increase in caveolae, prominin-2 is lost from the basal plasma membrane. In contrast, prominin-2 remains in the basolateral plasma membrane throughout OH pregnancy. Transmission electron microscopy showed that this membrane contained few caveolae throughout OH pregnancy. Our results indicate that prominin-2 prevents the formation of caveolae. We suggest the retention of prominin-2 in the basal plasma membrane during OH pregnancy prevents the formation of caveolae and is responsible for the retention of focal adhesions in this membrane, thereby contributing to the reduced implantation rate observed after such treatments.
Collapse
Affiliation(s)
- Samson N Dowland
- 1 Cell and Reproductive Biology Laboratory, School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Romanthi J Madawala
- 1 Cell and Reproductive Biology Laboratory, School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Connie E Poon
- 1 Cell and Reproductive Biology Laboratory, School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Laura A Lindsay
- 1 Cell and Reproductive Biology Laboratory, School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Christopher R Murphy
- 1 Cell and Reproductive Biology Laboratory, School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Pereira MFN, Fernandes SAF, Nascimento AR, Siu ER, Hess RA, Oliveira CA, Porto CS, Lazari MFM. Effects of the oestrogen receptor antagonist Fulvestrant on expression of genes that affect organization of the epididymal epithelium. Andrology 2014; 2:559-71. [PMID: 24782439 DOI: 10.1111/j.2047-2927.2014.00219.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/18/2014] [Accepted: 03/21/2014] [Indexed: 12/20/2022]
Abstract
The role of oestrogens in epididymal function is still unclear. Knockout of the oestrogen receptor ESR1 (Esr1(-/-) ) or treatment with the anti-oestrogen Fulvestrant affect epididymal milieu and sperm motility. We investigated the effect of in vivo treatment of rats with Fulvestrant on: (i) expression of genes that may be important for the architecture and function of the epididymal epithelium: prominins 1 and 2, metalloproteinase 7, claudin 7, beta-catenin and cadherin 13, and (ii) levels of oestradiol and testosterone, and expression of oestrogen and androgen receptors, in the initial segment (IS), caput, corpus and cauda epididymis. Fulvestrant (i) reduced gene expression of prominin 1 (variant 1) in the caput, reduced prominin 1 protein content in the caput epididymis and in the efferent ductules, and increased the localization of prominin 1 in microvilli of the caput and corpus; (ii) reduced gene expression of prominin 2 in the corpus and cauda epididymis; (iii) increased the metalloproteinase 7 content in the apical region of principal cells from IS/caput; (iv) reduced in the corpus epididymis, but increased in the efferent ductules, the cadherin 13 mRNA level; (v) reduced testosterone but increased oestradiol levels in the corpus and cauda; (vi) increased the androgen receptor protein content in all regions of the epididymis, and the oestrogen receptor GPER in the corpus and cauda epididymis. In conclusion, treatment with Fulvestrant induced regional-specific changes in hormonal and steroid receptor content, and affected expression of proteins important for epithelial organization and absorption/secretion. The mechanisms of oestrogen action may differ among epididymal regions, which may contribute to determine region-specific sperm functions.
Collapse
Affiliation(s)
- M F N Pereira
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Steiner AB, Kim T, Cabot V, Hudspeth AJ. Dynamic gene expression by putative hair-cell progenitors during regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A 2014; 111:E1393-401. [PMID: 24706895 PMCID: PMC3986164 DOI: 10.1073/pnas.1318692111] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hearing loss is most commonly caused by the destruction of mechanosensory hair cells in the ear. This condition is usually permanent: Despite the presence of putative hair-cell progenitors in the cochlea, hair cells are not naturally replenished in adult mammals. Unlike those of the mammalian ear, the progenitor cells of nonmammalian vertebrates can regenerate hair cells throughout life. The basis of this difference remains largely unexplored but may lie in molecular dissimilarities that affect how progenitors respond to hair-cell death. To approach this issue, we analyzed gene expression in hair-cell progenitors of the lateral-line system. We developed a transgenic line of zebrafish that expresses a red fluorescent protein in the presumptive hair-cell progenitors known as mantle cells. Fluorescence-activated cell sorting from the skins of transgenic larvae, followed by microarray-based expression analysis, revealed a constellation of transcripts that are specifically enriched in these cells. Gene expression analysis after hair-cell ablation uncovered a cohort of genes that are differentially regulated early in regeneration, suggesting possible roles in the response of progenitors to hair-cell death. These results provide a resource for studying hair-cell regeneration and the biology of sensory progenitor cells.
Collapse
Affiliation(s)
- Aaron B Steiner
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065
| | | | | | | |
Collapse
|
15
|
Glycolipid and Glycoprotein Expression During Neural Development. ADVANCES IN NEUROBIOLOGY 2014; 9:185-222. [DOI: 10.1007/978-1-4939-1154-7_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|