1
|
Moriyama M, Mori R, Hayakawa T, Moriyama H. FOXO3A Plays a Role in Wound Healing by Regulating Fibroblast Mitochondrial Dynamics. J Invest Dermatol 2025; 145:1489-1501.e4. [PMID: 39547393 DOI: 10.1016/j.jid.2024.10.600] [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: 11/28/2023] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
The skin plays a protective role against harmful environmental stress such as UV rays. Therefore, the skin is constantly exposed to potential injuries, and wound healing is a vital process for the survival of all higher organisms. Wound healing is dependent on aging and metabolic status at a whole-body level. Because the FOXO family plays a role in aging and metabolism, we investigated the molecular functions of FOXO3A in skin wound healing using FoxO3a-/- mice. We observed that FoxO3a-/- mice showed accelerated skin wound healing. During wound healing, more fibroblasts accumulated at the wound edges and migrated into the wound bed in FoxO3a-/- mice. Moreover, cell migration of dermal fibroblasts isolated from FoxO3a-/- mice was significantly induced. During the in vitro cell migration, we observed accelerated mitochondrial fragmentation and decreased oxygen consumption in the mitochondria of FoxO3a-/- fibroblasts. These changes were caused by the upregulation of mitochondrial Rho GTPase 1, which is an essential mediator of microtubule-based mitochondrial motility. Mitochondrial Rho GTPase 1 inhibition significantly attenuated cell migration, mitochondrial fragmentation, and mitochondrial recruitment to the leading edge of the cells. These data indicate that FOXO3A plays a crucial role in wound healing by regulating mitochondrial dynamics.
Collapse
Affiliation(s)
- Mariko Moriyama
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, Japan.
| | - Ryoichi Mori
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takao Hayakawa
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, Japan
| | - Hiroyuki Moriyama
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, Japan.
| |
Collapse
|
2
|
Liou Y, Moritoki N, Shindo T, Ishii T, Kishi K. Actin mesh in Re-epithelialization during skin regeneration in adult newt ( Cynops pyrrhogaster). Regen Ther 2025; 29:254-261. [PMID: 40230354 PMCID: PMC11995012 DOI: 10.1016/j.reth.2025.03.014] [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: 09/26/2024] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Studies have highlighted the role of actin cables in embryonic scarless wound healing across various species. However, evidence for similar structures in adult animals remains lacking. Adult newts, known for their exceptional skin regeneration capabilities, are considered promising models for postnatal human studies. This study investigated actin fiber formation and alignment during re-epithelialization in the Japanese fire-bellied newt (Cynops pyrrhogaster). Methods Full-thickness skin excisions were performed, and actin structures were analyzed using immunohistochemistry and electron microscopy. The role of actin in re-epithelialization was assessed by inhibiting its formation with cytochalasin B. Myosin, an interacting cytoskeletal molecule, was examined through immunohistochemistry, while E-cadherin, an adhesion molecule, was analyzed using both immunohistochemistry and electron microscopy. Results Rather than an actin cable a mesh-like actin structure, termed the "actin mesh," was identified via immunohistochemical analysis. The actin mesh developed alongside wound epidermis extension and disappeared following complete re-epithelialization. Inhibition of actin formation delayed re-epithelialization, although the overall healing process showed no significant difference from the control group. Immunohistochemistry revealed the presence of myosin II and E-cadherin alongside Filamentous actin. Electron microscopy further demonstrated actin-rich structures in the wound epidermis compared to normal skin and confirmed E-cadherin-mediated cell-cell adhesion in the wound area. Conclusions The actin mesh plays a critical role in facilitating rapid re-epithelialization in adult newts, presenting a valuable model for studying scarless wound healing in adult organisms. The involvement of interacting molecules such as myosin and E-cadherin provides insights into the underlying mechanisms of this process. This model offers potential applications for addressing intractable wounds in humans.
Collapse
Affiliation(s)
- Yu Liou
- Department of Medical Education, Linkou Chang Gung Memorial Hospital, No.5, Fusing St., Gueishan Township, Taoyuan County 333, Taiwan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinanomachi 35, Tokyo 160-8582, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Shinanomachi 35, Tokyo 160-8582, Japan
| | - Tatsuyuki Ishii
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinanomachi 35, Tokyo 160-8582, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinanomachi 35, Tokyo 160-8582, Japan
| |
Collapse
|
3
|
Scepanovic G, Balaghi N, Rothenberg KE, Fernandez-Gonzalez R. mTor limits autophagy to facilitate cell volume expansion and rapid wound repair in Drosophila embryos. Dev Cell 2025; 60:1400-1410.e3. [PMID: 39824179 DOI: 10.1016/j.devcel.2024.12.039] [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: 10/03/2023] [Revised: 07/16/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Embryonic wounds repair rapidly, with no inflammation or scarring. Embryonic wound healing is driven by collective cell movements facilitated by the increase in the volume of the cells adjacent to the wound. The mechanistic target of rapamycin (mTor) complex 1 (TORC1) is associated with cell growth. We found that disrupting TORC1 signaling in Drosophila embryos prevented cell volume increases and slowed down wound repair. Catabolic processes, such as autophagy, can inhibit cell growth. Five-dimensional microscopy demonstrated that the number of autophagosomes decreased during wound repair, suggesting that autophagy must be tightly regulated for rapid wound healing. mTor inhibition increased autophagy, and activating autophagy prevented cell volume expansion and slowed down wound closure. Finally, reducing autophagy in embryos with disrupted TORC1 signaling rescued cell volume changes and rapid wound repair. Together, our results show that TORC1 activation upon wounding negatively regulates autophagy, allowing cells to increase their volumes to facilitate rapid wound healing.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Negar Balaghi
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Katheryn E Rothenberg
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
4
|
Biscari G, Sanz Del Olmo N, Palumbo FS, Gaglio R, Garofalo G, Pitarresi G, Fiorica C, Malkoch M. Antimicrobial NIR-Responsive Hydrogels Based on Gellan Gum and Bis-MPA Polyester Dendrimers. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22448-22463. [PMID: 40193530 PMCID: PMC12012787 DOI: 10.1021/acsami.5c02386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025]
Abstract
In this study, a near-infrared (NIR)-responsive hydrogel based on ethylenediamine (EDA)-functionalized Gellan Gum was developed through a simple preparation method. This hydrogel incorporates in situ synthesized polydopamine (pDA) and was loaded with first- and second-generation antimicrobial bis-MPA polyester dendrimers (TMP-G1-[Cys]6 and MP-G2-[Cys]12), bearing cysteamine hydrochloride as peripheral functional groups. The intrinsic ability of pDA to scavenge reactive oxygen species (ROS) and convert NIR light at 810 nm into heat imparted radical scavenging activity and photothermal properties to the systems. It has been demonstrated that, due to the noncovalent interactions with both GG-EDA and pDA, dendrimers are retained differently within the sample depending on their molecular weight and the number of terminal positive charges. This difference in retention influences their antimicrobial activity against Pseudomonas aeruginosa and Staphylococcus aureus. In particular, it has been shown that the NIR-induced photothermal effect plays a crucial role in triggering the activity of the sample loaded with the most retained dendrimer, which possesses the highest number of terminal positive charges. The high physiological fluid absorption capacity makes these materials ideal for wound exudate management. In addition, their resistance to hydrolytic degradation can be exploited to reduce the frequency of dressing changes, potentially improving patient comfort. The dendrimer-loaded samples demonstrated low cytotoxicity toward human fetal dermal mesenchymal stromal cells (FD-MSCs) and human epidermal keratinocytes (HaCaT). These findings suggest that GG-EDA@pDA+TMP-G1-[Cys]6 or TMP-G2-[Cys]12 could be promising candidates for the treatment of infected skin wounds.
Collapse
Affiliation(s)
- Giuseppina Biscari
- Department
of Biological Chemical and Pharmaceutical Science and Technology (STEBICEF), University of Palermo, Via Archirafi 30-32, Palermo 90123, Italy
| | - Natalia Sanz Del Olmo
- School
of Engineering Sciences in Chemistry, Biotechnology and Health (CBH),
Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Teknikringen 56, Stockholm SE-100 44, Sweden
| | - Fabio S. Palumbo
- Department
of Biological Chemical and Pharmaceutical Science and Technology (STEBICEF), University of Palermo, Via Archirafi 30-32, Palermo 90123, Italy
| | - Raimondo Gaglio
- Department
of Agricultural, Food and Forest Sciences (SAAF), Università degli Studi di Palermo, Viale delle Scienze, Palermo 90128, Italy
| | - Giuliana Garofalo
- Department
of Agricultural, Food and Forest Sciences (SAAF), Università degli Studi di Palermo, Viale delle Scienze, Palermo 90128, Italy
| | - Giovanna Pitarresi
- Department
of Biological Chemical and Pharmaceutical Science and Technology (STEBICEF), University of Palermo, Via Archirafi 30-32, Palermo 90123, Italy
| | - Calogero Fiorica
- Department
of Biological Chemical and Pharmaceutical Science and Technology (STEBICEF), University of Palermo, Via Archirafi 30-32, Palermo 90123, Italy
| | - Michael Malkoch
- School
of Engineering Sciences in Chemistry, Biotechnology and Health (CBH),
Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Teknikringen 56, Stockholm SE-100 44, Sweden
| |
Collapse
|
5
|
She Y, Wu P, Wan W, Liu H, Liu R, Wang T, Wang M, Shen L, Yang Y, Huang X, Zhang X, Tian Y, Zhang K. Polysaccharides, proteins and DNA based stimulus responsive hydrogels promoting wound healing and repair: A review. Int J Biol Macromol 2025; 304:140961. [PMID: 39952504 DOI: 10.1016/j.ijbiomac.2025.140961] [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: 12/12/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The healing of various wounds remains a serious challenge in the medical field, hydrogel has high hydrophilicity and biocompatibility due to its unique network structure, which shows a strong advantage in the field of wound healing. Stimulus responsive hydrogels are particularly effective,which can control the material properties according to the external stimulus source, and provide more targeted treatment for different wounds. Here, we review physiological mechanisms of wound healing and the relationship between polysaccharides, proteins and DNA based stimulus responsive hydrogels and wound healing, materials commonly used of polysaccharides, proteins and DNA based stimulus responsive hydrogels, mechanisms of stimulus responsive hydrogels formation and network structure types, common properties of polysaccharides, proteins and DNA based stimulus responsive hydrogels for promoting wound healing and discuss their applications in medicine. Finally, the limitations and application prospects of polysaccharides, proteins and DNA based stimulus responsive hydrogels were discussed and evaluated. The review focuses on the biomedical use of polysaccharides, proteins and DNA based stimulus responsive hydrogels in wound healing and repair, and provides insights for the development of clinical related materials.
Collapse
Affiliation(s)
- Yumo She
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, China
| | - He Liu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Ruonan Liu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Mengyao Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Xiaoyue Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China
| | - Ye Tian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; Foshan Graduate School of Innovation, Northeastern University, Foshan 528300, China.
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
6
|
Huang YT, Calvi BR. Activation of a Src-JNK pathway in unscheduled endocycling cells of the Drosophila wing disc induces a chronic wounding response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642788. [PMID: 40161657 PMCID: PMC11952448 DOI: 10.1101/2025.03.12.642788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The endocycle is a specialized cell cycle during which cells undergo repeated G / S phases to replicate DNA without division, leading to large polyploid cells. The transition from a mitotic cycle to an endocycle can be triggered by various stresses, which results in unscheduled, or induced endocycling cells (iECs). While iECs can be beneficial for wound healing, they can also be detrimental by impairing tissue growth or promoting cancer. However, the regulation of endocycling and its role in tissue growth remain poorly understood. Using the Drosophila wing disc as a model, we previously demonstrated that iEC growth is arrested through a Jun N-Terminal Kinase (JNK)-dependent, reversible senescence-like response. However, it remains unclear how JNK is activated in iECs and how iECs impact overall tissue structure. In this study, we performed a genetic screen and identified the Src42A-Shark-Slpr pathway as an upstream regulator of JNK in iECs, leading to their senescence-like arrest. We found that tissues recognize iECs as wounds, releasing wound-related signals that induce a JNK-dependent developmental delay. Similar to wound closure, this response triggers Src-JNK-mediated actomyosin remodeling, yet iECs persist rather than being eliminated. Our findings suggest that the tissue response to iECs shares key signaling and cytoskeletal regulatory mechanisms with wound healing and dorsal closure, a developmental process during Drosophila embryogenesis. However, because iECs are retained within the tissue, they create a unique system that may serve as a model for studying chronic wounds and tumor progression.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
| | - Brian R. Calvi
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, 46202 USA
| |
Collapse
|
7
|
Ganguly A, Nanda S, Mandi M, Das K, Rajak P. Exploring the ameliorative potential of rutin against High-Sucrose Diet-induced oxidative stress and reproductive toxicity in Drosophila melanogaster. Reprod Toxicol 2024; 130:108742. [PMID: 39510200 DOI: 10.1016/j.reprotox.2024.108742] [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: 09/17/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Sucrose is a vital ingredient in numerous food items consumed regularly. However, exposure to excessive sucrose for a prolonged period can promote health issues. The reproductive system has a delicate physiology that can be targeted by various chemical stressors, including sucrose. Hence, the present in vivo study aims to unveil the impacts of High-Sucrose Diet (HSD) on the reproductive fitness of Drosophila melanogaster. In addition, the present work has also assessed the protective potential of a bioactive compound, rutin, against it. Here, first instar larvae were exposed to HSD (30 %) alone and in combination with rutin (100-300 µM) till their adult stage. HSD disturbed sex comb morphology in adult males, while fecundity and hatchability of eggs in females. Moreover, HSD triggered gonadal ROS production, oxidative stress, and modulated endogenous antioxidants such as SOD, catalase, and glutathione in both sexes. Nuclear fragmentation and tissue injuries, along with protein and lipid oxidation, were also apparent. Elevated levels of cytosolic Iron suggested an active Fenton reaction in adults. Further, HSD modulated the activities of reproductive and metabolic mediators, including vitellogenin, malate dehydrogenase, glucose-6-phosphate dehydrogenase, and angiotensin-converting enzymes that are critical to maintain the overall reproductive fitness. Interestingly, co-treatment with rutin, mainly at 200 µM, mitigated these adverse effects and restored reproductive fitness. The protective potential of rutin might be attributed to its ability to normalize redox homeostasis, reduce oxidative stress, and optimize critical enzymes involved in reproductive physiology. These findings suggest that rutin has potential therapeutic implications for counteracting the reproductive hazards induced by HSD.
Collapse
Affiliation(s)
- Abhratanu Ganguly
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Sayantani Nanda
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Moutushi Mandi
- Toxicology Research Unit, Department of Zoology, The University of Burdwan, Purba Bardhaman, West Bengal, India
| | - Kanchana Das
- Toxicology Research Unit, Department of Zoology, The University of Burdwan, Purba Bardhaman, West Bengal, India
| | - Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India.
| |
Collapse
|
8
|
Hunt M, Torres M, Bachar-Wikstrom E, Wikstrom JD. Cellular and molecular roles of reactive oxygen species in wound healing. Commun Biol 2024; 7:1534. [PMID: 39562800 DOI: 10.1038/s42003-024-07219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Wound healing is a highly coordinated spatiotemporal sequence of events involving several cell types and tissues. The process of wound healing requires strict regulation, and its disruption can lead to the formation of chronic wounds, which can have a significant impact on an individual's health as well as on worldwide healthcare expenditure. One essential aspect within the cellular and molecular regulation of wound healing pathogenesis is that of reactive oxygen species (ROS) and oxidative stress. Wounding significantly elevates levels of ROS, and an array of various reactive species are involved in modulating the wound healing process, such as through antimicrobial activities and signal transduction. However, as in many pathologies, ROS play an antagonistic pleiotropic role in wound healing, and can be a pathogenic factor in the formation of chronic wounds. Whilst advances in targeting ROS and oxidative stress have led to the development of novel pre-clinical therapeutic methods, due to the complex nature of ROS in wound healing, gaps in knowledge remain concerning the specific cellular and molecular functions of ROS in wound healing. In this review, we highlight current knowledge of these functions, and discuss the potential future direction of new studies, and how these pathways may be targeted in future pre-clinical studies.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
9
|
Shreesha L, Levin M. Stress sharing as cognitive glue for collective intelligences: A computational model of stress as a coordinator for morphogenesis. Biochem Biophys Res Commun 2024; 731:150396. [PMID: 39018974 PMCID: PMC11356093 DOI: 10.1016/j.bbrc.2024.150396] [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: 04/18/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Individual cells have numerous competencies in physiological and metabolic spaces. However, multicellular collectives can reliably navigate anatomical morphospace towards much larger, reliable endpoints. Understanding the robustness and control properties of this process is critical for evolutionary developmental biology, bioengineering, and regenerative medicine. One mechanism that has been proposed for enabling individual cells to coordinate toward specific morphological outcomes is the sharing of stress (where stress is a physiological parameter that reflects the current amount of error in the context of a homeostatic loop). Here, we construct and analyze a multiscale agent-based model of morphogenesis in which we quantitatively examine the impact of stress sharing on the ability to reach target morphology. We found that stress sharing improves the morphogenetic efficiency of multicellular collectives; populations with stress sharing reached anatomical targets faster. Moreover, stress sharing influenced the future fate of distant cells in the multi-cellular collective, enhancing cells' movement and their radius of influence, consistent with the hypothesis that stress sharing works to increase cohesiveness of collectives. During development, anatomical goal states could not be inferred from observation of stress states, revealing the limitations of knowledge of goals by an extern observer outside the system itself. Taken together, our analyses support an important role for stress sharing in natural and engineered systems that seek robust large-scale behaviors to emerge from the activity of their competent components.
Collapse
Affiliation(s)
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA, 02155, USA; Allen Discovery Center at Tufts University, Medford, MA, 02155, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Kahlon U, Ricca FD, Pillai SJ, Olivetta M, Tharp KM, Jao LE, Dudin O, McDonald K, Aydogan MG. A mitochondrial redox switch licenses the onset of morphogenesis in animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620733. [PMID: 39553983 PMCID: PMC11565760 DOI: 10.1101/2024.10.28.620733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Embryos undergo pre-gastrulation cleavage cycles to generate a critical cell mass before transitioning to morphogenesis. The molecular underpinnings of this transition have traditionally centered on zygotic chromatin remodeling and genome activation1,2, as their repression can prevent downstream processes of differentiation and organogenesis. Despite precedents that oxygen depletion can similarly suspend development in early embryos3-6, hinting at a pivotal role for oxygen metabolism in this transition, whether there is a bona fide chemical switch that licenses the onset of morphogenesis remains unknown. Here we discover that a mitochondrial oxidant acts as a metabolic switch to license the onset of animal morphogenesis. Concomitant with the instatement of mitochondrial membrane potential, we found a burst-like accumulation of mitochondrial superoxide (O2 -) during fly blastoderm formation. In vivo chemistry experiments revealed that an electron leak from site IIIQo at ETC Complex III is responsible for O2 - production. Importantly, depleting mitochondrial O2 - fully mimics anoxic conditions and, like anoxia, induces suspended animation prior to morphogenesis, but not after. Specifically, H2O2, and not ONOO-, NO, or HO•, can single-handedly account for this mtROS-based response. We demonstrate that depleting mitochondrial O2 - similarly prevents the onset of morphogenetic events in vertebrate embryos and ichthyosporea, close relatives of animals. We postulate that such redox-based metabolic licensing of morphogenesis is an ancient trait of holozoans that couples the availability of oxygen to development, conserved from early-diverging animal relatives to vertebrates.
Collapse
Affiliation(s)
- Updip Kahlon
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Touro College of Osteopathic Medicine, Touro University, USA
- These authors have contributed equally
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Dev. & Stem Cell Biology Graduate Program, University of California, San Francisco, USA
- These authors have contributed equally
| | - Saraswathi J. Pillai
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- These authors have contributed equally
| | - Marine Olivetta
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Switzerland
| | - Kevin M. Tharp
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, USA
| | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California, Davis, USA
| | - Omaya Dudin
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Switzerland
| | - Kent McDonald
- Electron Microscope Lab, University of California, Berkeley, USA
| | - Mustafa G. Aydogan
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Nutrition and Obesity Research Center, University of California, San Francisco, USA
| |
Collapse
|
11
|
Vicidomini C, Palumbo R, Moccia M, Roviello GN. Oxidative Processes and Xenobiotic Metabolism in Plants: Mechanisms of Defense and Potential Therapeutic Implications. J Xenobiot 2024; 14:1541-1569. [PMID: 39449425 PMCID: PMC11503355 DOI: 10.3390/jox14040084] [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: 08/08/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Plants are continuously exposed to environmental challenges, including pollutants, pesticides, and heavy metals, collectively termed xenobiotics. These substances induce oxidative stress by generating reactive oxygen species (ROS), which can damage cellular components such as lipids, proteins, and nucleic acids. To counteract this, plants have evolved complex metabolic pathways to detoxify and process these harmful compounds. Oxidative stress in plants primarily arises from the overproduction of hydrogen peroxide (H2O2), superoxide anions (O2•-), singlet oxygen (1O2), and hydroxyl radicals (•OH), by-products of metabolic activities such as photosynthesis and respiration. The presence of xenobiotics leads to a notable increase in ROS, which can result in cellular damage and metabolic disruption. To combat this, plants have developed a strong antioxidant defense mechanism that includes enzymatic antioxidants that work together to eliminate ROS, thereby reducing their harmful effects. In addition to enzymatic defenses, plants also synthesize various non-enzymatic antioxidants, including flavonoids, phenolic acids, and vitamins. These compounds effectively neutralize ROS and help regenerate other antioxidants, offering extensive protection against oxidative stress. The metabolism of xenobiotic substances in plants occurs in three stages: the first involves modification, which refers to the chemical alteration of xenobiotics to make them less harmful. The second involves conjugation, where the modified xenobiotics are combined with other substances to increase their solubility, facilitating their elimination from the plant. The third stage involves compartmentalization, which is the storage or isolation of conjugated xenobiotics in specific parts of the plant, helping to prevent damage to vital cellular functions. Secondary metabolites found in plants, such as alkaloids, terpenoids, and flavonoids, play a vital role in detoxification and the defense against oxidative stress. Gaining a deeper understanding of the oxidative mechanisms and the pathways of xenobiotic metabolism in plants is essential, as this knowledge can lead to the formulation of plant-derived strategies aimed at alleviating the effects of environmental pollution and enhancing human health by improving detoxification and antioxidant capabilities, as discussed in this review.
Collapse
Affiliation(s)
- Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Maria Moccia
- Institute of Crystallography, Italian National Council for Research (IC-CNR), Strada Provinciale 35d, 9, Montelibretti, 00010 Rome, Italy
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| |
Collapse
|
12
|
Qiu H, Sun Y, Wang X, Gong T, Su J, Shen J, Zhou J, Xia J, Wang H, Meng X, Fu G, Zhang D, Jiang C, Liang P. Lamin A/C deficiency-mediated ROS elevation contributes to pathogenic phenotypes of dilated cardiomyopathy in iPSC model. Nat Commun 2024; 15:7000. [PMID: 39143095 PMCID: PMC11324749 DOI: 10.1038/s41467-024-51318-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.
Collapse
Affiliation(s)
- Hangyuan Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yaxun Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Xiaochen Wang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Tingyu Gong
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jun Su
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jiaxi Shen
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jingjun Zhou
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wang
- Prenatal Diagnosis Center, Hangzhou Women's Hospital, Hangzhou, China
| | - Xiangfu Meng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China.
| | - Ping Liang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Ben-Hur S, Sernik S, Afar S, Kolpakova A, Politi Y, Gal L, Florentin A, Golani O, Sivan E, Dezorella N, Morgenstern D, Pietrokovski S, Schejter E, Yacobi-Sharon K, Arama E. Egg multivesicular bodies elicit an LC3-associated phagocytosis-like pathway to degrade paternal mitochondria after fertilization. Nat Commun 2024; 15:5715. [PMID: 38977659 PMCID: PMC11231261 DOI: 10.1038/s41467-024-50041-5] [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: 09/20/2023] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Mitochondria are maternally inherited, but the mechanisms underlying paternal mitochondrial elimination after fertilization are far less clear. Using Drosophila, we show that special egg-derived multivesicular body vesicles promote paternal mitochondrial elimination by activating an LC3-associated phagocytosis-like pathway, a cellular defense pathway commonly employed against invading microbes. Upon fertilization, these egg-derived vesicles form extended vesicular sheaths around the sperm flagellum, promoting degradation of the sperm mitochondrial derivative and plasma membrane. LC3-associated phagocytosis cascade of events, including recruitment of a Rubicon-based class III PI(3)K complex to the flagellum vesicular sheaths, its activation, and consequent recruitment of Atg8/LC3, are all required for paternal mitochondrial elimination. Finally, lysosomes fuse with strings of large vesicles derived from the flagellum vesicular sheaths and contain degrading fragments of the paternal mitochondrial derivative. Given reports showing that in some mammals, the paternal mitochondria are also decorated with Atg8/LC3 and surrounded by multivesicular bodies upon fertilization, our findings suggest that a similar pathway also mediates paternal mitochondrial elimination in other flagellated sperm-producing organisms.
Collapse
Affiliation(s)
- Sharon Ben-Hur
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shoshana Sernik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sara Afar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Alina Kolpakova
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yoav Politi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Anat Florentin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ehud Sivan
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Nili Dezorella
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalised Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Shmuel Pietrokovski
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Yacobi-Sharon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
14
|
Uttekar B, Verma RK, Tomer D, Rikhy R. Mitochondrial morphology dynamics and ROS regulate apical polarity and differentiation in Drosophila follicle cells. Development 2024; 151:dev201732. [PMID: 38345270 PMCID: PMC7616099 DOI: 10.1242/dev.201732] [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: 03/10/2023] [Accepted: 01/23/2024] [Indexed: 03/01/2024]
Abstract
Mitochondrial morphology dynamics regulate signaling pathways during epithelial cell formation and differentiation. The mitochondrial fission protein Drp1 affects the appropriate activation of EGFR and Notch signaling-driven differentiation of posterior follicle cells in Drosophila oogenesis. The mechanisms by which Drp1 regulates epithelial polarity during differentiation are not known. In this study, we show that Drp1-depleted follicle cells are constricted in early stages and present in multiple layers at later stages with decreased levels of apical polarity protein aPKC. These defects are suppressed by additional depletion of mitochondrial fusion protein Opa1. Opa1 depletion leads to mitochondrial fragmentation and increased reactive oxygen species (ROS) in follicle cells. We find that increasing ROS by depleting the ROS scavengers, mitochondrial SOD2 and catalase also leads to mitochondrial fragmentation. Further, the loss of Opa1, SOD2 and catalase partially restores the defects in epithelial polarity and aPKC, along with EGFR and Notch signaling in Drp1-depleted follicle cells. Our results show a crucial interaction between mitochondrial morphology, ROS generation and epithelial cell polarity formation during the differentiation of follicle epithelial cells in Drosophila oogenesis.
Collapse
Affiliation(s)
- Bhavin Uttekar
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Rahul Kumar Verma
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Darshika Tomer
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Richa Rikhy
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| |
Collapse
|
15
|
Ding C, Liu X, Zhang S, Sun S, Yang J, Chai G, Wang N, Ma S, Ding Q, Liu W. Multifunctional hydrogel bioscaffolds based on polysaccharide to promote wound healing: A review. Int J Biol Macromol 2024; 259:129356. [PMID: 38218300 DOI: 10.1016/j.ijbiomac.2024.129356] [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: 09/14/2023] [Revised: 12/24/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
Various types of skin wounds pose challenges in terms of healing and susceptibility to infection, which can have a significant impact on physical and mental well-being, and in severe cases, may result in amputation. Conventional wound dressings often fail to provide optimal support for these wounds, thereby impeding the healing process. As a result, there has been considerable interest in the development of multifunctional polymer matrix hydrogel scaffolds for wound healing. This review offers a comprehensive review of the characteristics of polysaccharide-based hydrogel scaffolds, as well as their applications in different types of wounds. Additionally, it evaluates the advantages and disadvantages associated with various types of multifunctional polymer and polysaccharide-based hydrogel scaffolds. The objective is to provide a theoretical foundation for the utilization of multifunctional hydrogel scaffolds in promoting wound healing.
Collapse
Affiliation(s)
- Chuanbo Ding
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Xinglong Liu
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Shuai Zhang
- Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- Jilin Agricultural University, Changchun 130118, China
| | - Jiali Yang
- Jilin Agricultural University, Changchun 130118, China
| | - Guodong Chai
- Jilin Agricultural University, Changchun 130118, China
| | - Ning Wang
- Jilin Agricultural University, Changchun 130118, China
| | - Shuang Ma
- Jilin Agricultural University, Changchun 130118, China
| | - Qiteng Ding
- Jilin Agricultural University, Changchun 130118, China; Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Changchun 130118, China.
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China.
| |
Collapse
|
16
|
Pérez-Montes C, Jiménez-Cubides JP, Velasco A, Arévalo R, Santos-Ledo A, García-Macia M. REDOX Balance in Oligodendrocytes Is Important for Zebrafish Visual System Regeneration. Antioxidants (Basel) 2023; 12:2026. [PMID: 38136146 PMCID: PMC10740785 DOI: 10.3390/antiox12122026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Zebrafish (Danio rerio) present continuous growth and regenerate many parts of their body after an injury. Fish oligodendrocytes, microglia and astrocytes support the formation of new connections producing effective regeneration of the central nervous system after a lesion. To understand the role of oligodendrocytes and the signals that mediate regeneration, we use the well-established optic nerve (ON) crush model. We also used sox10 fluorescent transgenic lines to label fully differentiated oligodendrocytes. To quench the effect of reactive oxygen species (ROS), we used the endogenous antioxidant melatonin. Using these tools, we measured ROS production by flow cytometry and explored the regeneration of the optic tectum (OT), the response of oligodendrocytes and their mitochondria by confocal microscopy and Western blot. ROS are produced by oligodendrocytes 3 h after injury and JNK activity is triggered. Concomitantly, there is a decrease in the number of fully differentiated oligodendrocytes in the OT and in their mitochondrial population. By 24 h, oligodendrocytes partially recover. Exposure to melatonin blocks the changes observed in these oligodendrocytes at 3 h and increases their number and their mitochondrial populations after 24 h. Melatonin also blocks JNK upregulation and induces aberrant neuronal differentiation in the OT. In conclusion, a proper balance of ROS is necessary during visual system regeneration and exposure to melatonin has a detrimental impact.
Collapse
Affiliation(s)
- Cristina Pérez-Montes
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.P.-M.); (J.P.J.-C.); (A.V.); (R.A.)
- Department of Human Anatomy and Histology, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Jhoana Paola Jiménez-Cubides
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.P.-M.); (J.P.J.-C.); (A.V.); (R.A.)
| | - Almudena Velasco
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.P.-M.); (J.P.J.-C.); (A.V.); (R.A.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Cell Biology and Pathology, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Rosario Arévalo
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.P.-M.); (J.P.J.-C.); (A.V.); (R.A.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Cell Biology and Pathology, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Adrián Santos-Ledo
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.P.-M.); (J.P.J.-C.); (A.V.); (R.A.)
- Department of Human Anatomy and Histology, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Marina García-Macia
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Department of Biochemistry and Molecular Biology, Universidad de Salamanca, 37007 Salamanca, Spain
- Centre for Biomedical Investigations Network on Frailty and Ageing (CIBERFES), 28029 Madrid, Spain
| |
Collapse
|
17
|
Hunt M, Torres M, Bachar-Wikström E, Wikström JD. Multifaceted roles of mitochondria in wound healing and chronic wound pathogenesis. Front Cell Dev Biol 2023; 11:1252318. [PMID: 37771375 PMCID: PMC10523588 DOI: 10.3389/fcell.2023.1252318] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Mitochondria are intracellular organelles that play a critical role in numerous cellular processes including the regulation of metabolism, cellular stress response, and cell fate. Mitochondria themselves are subject to well-orchestrated regulation in order to maintain organelle and cellular homeostasis. Wound healing is a multifactorial process that involves the stringent regulation of several cell types and cellular processes. In the event of dysregulated wound healing, hard-to-heal chronic wounds form and can place a significant burden on healthcare systems. Importantly, treatment options remain limited owing to the multifactorial nature of chronic wound pathogenesis. One area that has received more attention in recent years is the role of mitochondria in wound healing. With regards to this, current literature has demonstrated an important role for mitochondria in several areas of wound healing and chronic wound pathogenesis including metabolism, apoptosis, and redox signalling. Additionally, the influence of mitochondrial dynamics and mitophagy has also been investigated. However, few studies have utilised patient tissue when studying mitochondria in wound healing, instead using various animal models. In this review we dissect the current knowledge of the role of mitochondria in wound healing and discuss how future research can potentially aid in the progression of wound healing research.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Rothenberg KE, Chen Y, McDonald JA, Fernandez-Gonzalez R. Rap1 coordinates cell-cell adhesion and cytoskeletal reorganization to drive collective cell migration in vivo. Curr Biol 2023:S0960-9822(23)00603-6. [PMID: 37244252 DOI: 10.1016/j.cub.2023.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/16/2023] [Accepted: 05/04/2023] [Indexed: 05/29/2023]
Abstract
Collective cell movements contribute to tissue development and repair and spread metastatic disease. In epithelia, cohesive cell movements require reorganization of adherens junctions and the actomyosin cytoskeleton. However, the mechanisms that coordinate cell-cell adhesion and cytoskeletal remodeling during collective cell migration in vivo are unclear. We investigated the mechanisms of collective cell migration during epidermal wound healing in Drosophila embryos. Upon wounding, the cells adjacent to the wound internalize cell-cell adhesion molecules and polarize actin and the motor protein non-muscle myosin II to form a supracellular cable around the wound that coordinates cell movements. The cable anchors at former tricellular junctions (TCJs) along the wound edge, and TCJs are reinforced during wound closure. We found that the small GTPase Rap1 was necessary and sufficient for rapid wound repair. Rap1 promoted myosin polarization to the wound edge and E-cadherin accumulation at TCJs. Using embryos expressing a mutant form of the Rap1 effector Canoe/Afadin that cannot bind Rap1, we found that Rap1 signals through Canoe for adherens junction remodeling, but not for actomyosin cable assembly. Instead, Rap1 was necessary and sufficient for RhoA/Rho1 activation at the wound edge. The RhoGEF Ephexin localized to the wound edge in a Rap1-dependent manner, and Ephexin was necessary for myosin polarization and rapid wound repair, but not for E-cadherin redistribution. Together, our data show that Rap1 coordinates the molecular rearrangements that drive embryonic wound healing, promoting actomyosin cable assembly through Ephexin-Rho1, and E-cadherin redistribution through Canoe, thus enabling rapid collective cell migration in vivo.
Collapse
Affiliation(s)
- Katheryn E Rothenberg
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Yujun Chen
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | | | - Rodrigo Fernandez-Gonzalez
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
19
|
Sokac AM, Biel N, De Renzis S. Membrane-actin interactions in morphogenesis: Lessons learned from Drosophila cellularization. Semin Cell Dev Biol 2023; 133:107-122. [PMID: 35396167 PMCID: PMC9532467 DOI: 10.1016/j.semcdb.2022.03.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/12/2023]
Abstract
During morphogenesis, changes in the shapes of individual cells are harnessed to mold an entire tissue. These changes in cell shapes require the coupled remodeling of the plasma membrane and underlying actin cytoskeleton. In this review, we highlight cellularization of the Drosophila embryo as a model system to uncover principles of how membrane and actin dynamics are co-regulated in space and time to drive morphogenesis.
Collapse
Affiliation(s)
- Anna Marie Sokac
- Department of Cell and Developmental Biology, University of Illinois at Urbana Champaign, Urbana, IL 61801, USA; Graduate Program in Integrative and Molecular Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Natalie Biel
- Department of Cell and Developmental Biology, University of Illinois at Urbana Champaign, Urbana, IL 61801, USA; Graduate Program in Integrative and Molecular Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stefano De Renzis
- European Molecular Biology Laboratory Heidelberg, 69117 Heidelberg, Germany
| |
Collapse
|
20
|
Contractile and expansive actin networks in Drosophila: Developmental cell biology controlled by network polarization and higher-order interactions. Curr Top Dev Biol 2023; 154:99-129. [PMID: 37100525 DOI: 10.1016/bs.ctdb.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
Actin networks are central to shaping and moving cells during animal development. Various spatial cues activate conserved signal transduction pathways to polarize actin network assembly at sub-cellular locations and to elicit specific physical changes. Actomyosin networks contract and Arp2/3 networks expand, and to affect whole cells and tissues they do so within higher-order systems. At the scale of tissues, actomyosin networks of epithelial cells can be coupled via adherens junctions to form supracellular networks. Arp2/3 networks typically integrate with distinct actin assemblies, forming expansive composites which act in conjunction with contractile actomyosin networks for whole-cell effects. This review explores these concepts using examples from Drosophila development. First, we discuss the polarized assembly of supracellular actomyosin cables which constrict and reshape epithelial tissues during embryonic wound healing, germ band extension, and mesoderm invagination, but which also form physical borders between tissue compartments at parasegment boundaries and during dorsal closure. Second, we review how locally induced Arp2/3 networks act in opposition to actomyosin structures during myoblast cell-cell fusion and cortical compartmentalization of the syncytial embryo, and how Arp2/3 and actomyosin networks also cooperate for the single cell migration of hemocytes and the collective migration of border cells. Overall, these examples show how the polarized deployment and higher-order interactions of actin networks organize developmental cell biology.
Collapse
|
21
|
Wang G, Yang F, Zhou W, Xiao N, Luo M, Tang Z. The initiation of oxidative stress and therapeutic strategies in wound healing. Biomed Pharmacother 2023; 157:114004. [PMID: 36375308 DOI: 10.1016/j.biopha.2022.114004] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
When the production of reactive oxygen species (ROS) is overloaded surpassing the capacity of the reductive rheostat, mammalian cells undergo a series of oxidative damage termed oxidative stress (OS). This phenomenon is ubiquitously detected in many human pathological conditions. Wound healing program implicates continuous neovascularization, cell proliferation, and wound remodeling. Increasing evidence indicates that reactive oxygen species (ROS) have profound impacts on the wound healing process through regulating a series of the physiological and pathological program including inflammatory response, cell proliferation, angiogenesis, granulation as well as extracellular matrix formation. In most pathological wound healing processes, excessive ROS exerts a negative role on the wound healing process. Interestingly, the moderate increase of ROS levels is beneficial in killing bacteria at the wound site, which creates a sterile niche for revascularization. In this review, we discussed the physiological rhythms of wound healing and the role of ROS in this progress, aim to explore the potential manipulation of OS as a promising therapeutic avenue.
Collapse
Affiliation(s)
- Gang Wang
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China; Drug Discovery Research Center, Southwest Medical University, Luzhou, China; Department of Pharmacology, college of Pharmacy, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Drug Metabolism, Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China, Chongqing, China
| | - Feifei Yang
- Department of Pharmacology, college of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Weiying Zhou
- Department of Pharmacology, college of Pharmacy, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Drug Metabolism, Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China, Chongqing, China
| | - Nanyang Xiao
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Mao Luo
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China; Drug Discovery Research Center, Southwest Medical University, Luzhou, China.
| | - Zonghao Tang
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China; Drug Discovery Research Center, Southwest Medical University, Luzhou, China; Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Xu S, Li S, Bjorklund M, Xu S. Mitochondrial fragmentation and ROS signaling in wound response and repair. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:38. [PMID: 36451031 PMCID: PMC9712903 DOI: 10.1186/s13619-022-00141-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022]
Abstract
Mitochondria are organelles that serve numerous critical cellular functions, including energy production, Ca2+ homeostasis, redox signaling, and metabolism. These functions are intimately linked to mitochondrial morphology, which is highly dynamic and capable of rapid and transient changes to alter cellular functions in response to environmental cues and cellular demands. Mitochondrial morphology and activity are critical for various physiological processes, including wound healing. In mammals, wound healing is a complex process that requires coordinated function of multiple cell types and progresses in partially overlapping but distinct stages: hemostasis and inflammation, cell proliferation and migration, and tissue remodeling. The repair process at the single-cell level forms the basis for wound healing and regeneration in tissues. Recent findings reveal that mitochondria fulfill the intensive energy demand for wound repair and aid wound closure by cytoskeleton remodeling via morphological changes and mitochondrial reactive oxygen species (mtROS) signaling. In this review, we will mainly elucidate how wounding induces changes in mitochondrial morphology and activity and how these changes, in turn, contribute to cellular wound response and repair.
Collapse
Affiliation(s)
- Shiqi Xu
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Shiyao Li
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Mikael Bjorklund
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China
| | - Suhong Xu
- Zhejiang University-University of Edinburgh Institute and International Biomedicine-X Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 718 East Haizhou Rd., Haining, 314400, Zhejiang, China.
- Center for Stem Cell and Regenerative Medicine and Department of Burn and wound repair of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
23
|
Serras F. The sooner, the better: ROS, kinases and nutrients at the onset of the damage response in Drosophila. Front Cell Dev Biol 2022; 10:1047823. [PMID: 36353511 PMCID: PMC9637634 DOI: 10.3389/fcell.2022.1047823] [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: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
One of the main topics in regeneration biology is the nature of the early signals that trigger the damage response. Recent advances in Drosophila point to the MAP3 kinase Ask1 as a molecular hub that integrates several signals at the onset of regeneration. It has been discovered that reactive oxygen species (ROS) produced in damaged imaginal discs and gut epithelia will activate the MAP3 kinase Ask1. Severely damaged and apoptotic cells produce an enormous amount of ROS, which ensures their elimination by activating Ask1 and in turn the pro-apoptotic function of JNK. However, this creates an oxidative stress environment with beneficial effects that is sensed by neighboring healthy cells. This environment, in addition to the Pi3K/Akt nutrient sensing pathway, can be integrated into Ask1 to launch regeneration. Ultimately the activity of Ask1 depends on these and other inputs and modulates its signaling to achieve moderate levels of p38 and low JNK signaling and thus promote survival and regeneration. This model based on the dual function of Ask1 for early response to damage is discussed here.
Collapse
Affiliation(s)
- Florenci Serras
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Li Z, Tang Y, Cai J, Wu S, Song F. MPP7 as a Novel Biomarker of Esophageal Cancer: MPP7 Knockdown Inhibits Esophageal Cancer Cell Migration and Invasion. Life (Basel) 2022; 12:life12091381. [PMID: 36143417 PMCID: PMC9501138 DOI: 10.3390/life12091381] [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: 08/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
MAGUK p55 scaffold protein 7 (MPP7) is a member of the stardust family of membrane-associated guanosine kinase protein P55 and plays a role in the establishment of epithelial cell polarity. However, its potential implication in human esophageal cancer is unclear. In this study, we investigated the expression profile of MPP7 and its functional impact on esophagus cancer. Expression analyses of immunohistochemical microarrays with survival and prognostic information of 103 patients with esophageal cancer demonstrated that MPP7 was overexpressed in 52 patients, who showed poor survival rates. The transcriptional expression of MPP7 in esophageal cancer in TCGA database increased successively from normal epithelial, to esophageal adenocarcinoma, to esophageal squamous cell carcinoma. Transcriptome sequencing after MPP7 knockdown in esophageal carcinoma cells showed that wound-healing-associated proteins were down-regulated, and the TGF-β pathway was one of the important signaling pathways. A loss-of-function study showed that the knockdown of MPP7 inhibited cell migration and invasion. These results could be verified in a model of tumor cells injected into the tail vein and subcutaneous tumor of nude mice. Herein, our results indicated that MPP7 could have an oncogenic role in human esophagus cancer, thus demonstrating its potential as a novel biomarker for the diagnosis and/or treatment of esophagus cancer.
Collapse
Affiliation(s)
- Zhaodong Li
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 1#Yixue Yuan Road, Yuzhong District, Chongqing 400016, China
| | - Yongyao Tang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 1#Yixue Yuan Road, Yuzhong District, Chongqing 400016, China
| | - Jing Cai
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 1#Yixue Yuan Road, Yuzhong District, Chongqing 400016, China
| | - Shunlong Wu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fangzhou Song
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 1#Yixue Yuan Road, Yuzhong District, Chongqing 400016, China
- Correspondence: ; Tel.: +86-23-68485958
| |
Collapse
|
25
|
Qin S, Li B, Ming H, Nice EC, Zou B, Huang C. Harnessing redox signaling to overcome therapeutic-resistant cancer dormancy. Biochim Biophys Acta Rev Cancer 2022; 1877:188749. [PMID: 35716972 DOI: 10.1016/j.bbcan.2022.188749] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023]
Abstract
Dormancy occurs when cells preserve viability but stop proliferating, which is considered an important cause of tumor relapse, which may occur many years after clinical remission. Since the life cycle of dormant cancer cells is affected by both intracellular and extracellular factors, gene mutation or epigenetic regulation of tumor cells may not fully explain the mechanisms involved. Recent studies have indicated that redox signaling regulates the formation, maintenance, and reactivation of dormant cancer cells by modulating intracellular signaling pathways and the extracellular environment, which provides a molecular explanation for the life cycle of dormant tumor cells. Indeed, redox signaling regulates the onset of dormancy by balancing the intrinsic pathways, the extrinsic environment, and the response to therapy. In addition, redox signaling sustains dormancy by managing stress homeostasis, maintaining stemness and immunogenic equilibrium. However, studies on dormancy reactivation are still limited, partly explained by redox-mediated activation of lipid metabolism and the transition from the tumor microenvironment to inflammation. Encouragingly, several drug combination strategies based on redox biology are currently under clinical evaluation. Continuing to gain an in-depth understanding of redox regulation and develop specific methods targeting redox modification holds the promise to accelerate the development of strategies to treat dormant tumors and benefit cancer patients.
Collapse
Affiliation(s)
- Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Bingwen Zou
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China.
| |
Collapse
|
26
|
Mitochondria preserve an autarkic one-carbon cycle to confer growth-independent cancer cell migration and metastasis. Nat Commun 2022; 13:2699. [PMID: 35577770 PMCID: PMC9110368 DOI: 10.1038/s41467-022-30363-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/22/2022] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the most common cause of death in cancer patients. Canonical drugs target mainly the proliferative capacity of cancer cells, which leaves slow-proliferating, persistent cancer cells unaffected. Metabolic determinants that contribute to growth-independent functions are still poorly understood. Here we show that antifolate treatment results in an uncoupled and autarkic mitochondrial one-carbon (1C) metabolism during cytosolic 1C metabolism impairment. Interestingly, antifolate dependent growth-arrest does not correlate with decreased migration capacity. Therefore, using methotrexate as a tool compound allows us to disentangle proliferation and migration to profile the metabolic phenotype of migrating cells. We observe that increased serine de novo synthesis (SSP) supports mitochondrial serine catabolism and inhibition of SSP using the competitive PHGDH-inhibitor BI-4916 reduces cancer cell migration. Furthermore, we show that sole inhibition of mitochondrial serine catabolism does not affect primary breast tumor growth but strongly inhibits pulmonary metastasis. We conclude that mitochondrial 1C metabolism, despite being dispensable for proliferative capacities, confers an advantage to cancer cells by supporting their motility potential. Chemotherapeutic antifolates, such as methotrexate (MTX), impair cancer cell proliferation by inhibiting nucleotide synthesis. Here, the authors show that MTX sustains an autarkic mitochondrial one-carbon metabolism leading to serine synthesis to promote cancer cell migration and metastasis.
Collapse
|
27
|
Madan S, Uttekar B, Chowdhary S, Rikhy R. Mitochondria Lead the Way: Mitochondrial Dynamics and Function in Cellular Movements in Development and Disease. Front Cell Dev Biol 2022; 9:781933. [PMID: 35186947 PMCID: PMC8848284 DOI: 10.3389/fcell.2021.781933] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023] Open
Abstract
The dynamics, distribution and activity of subcellular organelles are integral to regulating cell shape changes during various physiological processes such as epithelial cell formation, cell migration and morphogenesis. Mitochondria are famously known as the powerhouse of the cell and play an important role in buffering calcium, releasing reactive oxygen species and key metabolites for various activities in a eukaryotic cell. Mitochondrial dynamics and morphology changes regulate these functions and their regulation is, in turn, crucial for various morphogenetic processes. In this review, we evaluate recent literature which highlights the role of mitochondrial morphology and activity during cell shape changes in epithelial cell formation, cell division, cell migration and tissue morphogenesis during organism development and in disease. In general, we find that mitochondrial shape is regulated for their distribution or translocation to the sites of active cell shape dynamics or morphogenesis. Often, key metabolites released locally and molecules buffered by mitochondria play crucial roles in regulating signaling pathways that motivate changes in cell shape, mitochondrial shape and mitochondrial activity. We conclude that mechanistic analysis of interactions between mitochondrial morphology, activity, signaling pathways and cell shape changes across the various cell and animal-based model systems holds the key to deciphering the common principles for this interaction.
Collapse
|
28
|
Sensing microbial infections in the Drosophila melanogaster genetic model organism. Immunogenetics 2022; 74:35-62. [DOI: 10.1007/s00251-021-01239-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022]
|
29
|
Xu J, Meng X, Yang Q, Zhang J, Hu W, Fu H, Chen JW, Ma W, Chisholm AD, Sun Q, Xu S. Redox-sensitive CDC-42 clustering promotes wound closure in C. elegans. Cell Rep 2021; 37:110040. [PMID: 34818546 DOI: 10.1016/j.celrep.2021.110040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 09/09/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022] Open
Abstract
Tissue damage induces immediate-early signals, activating Rho small GTPases to trigger actin polymerization essential for later wound repair. However, how tissue damage is sensed to activate Rho small GTPases locally remains elusive. Here, we found that wounding the C. elegans epidermis induces rapid relocalization of CDC-42 into plasma membrane-associated clusters, which subsequently recruits WASP/WSP-1 to trigger actin polymerization to close the wound. In addition, wounding induces a local transient increase and subsequent reduction of H2O2, which negatively regulates the clustering of CDC-42 and wound closure. CDC-42 CAAX motif-mediated prenylation and polybasic region-mediated cation-phospholipid interaction are both required for its clustering. Cysteine residues participate in intermolecular disulfide bonds to reduce membrane association and are required for negative regulation of CDC-42 clustering by H2O2. Collectively, our findings suggest that H2O2-regulated fine-tuning of CDC-42 localization can create a distinct biomolecular cluster that facilitates rapid epithelial wound repair after injury.
Collapse
Affiliation(s)
- Jingxiu Xu
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinan Meng
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang University-University of Edinburgh Institute, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qingxian Yang
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jianqin Zhang
- Department of Biochemistry and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Hu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongying Fu
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jack Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weirui Ma
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Andrew D Chisholm
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Qiming Sun
- Department of Biochemistry and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Suhong Xu
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang University-University of Edinburgh Institute, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
30
|
Scepanovic G, Hunter MV, Kafri R, Fernandez-Gonzalez R. p38-mediated cell growth and survival drive rapid embryonic wound repair. Cell Rep 2021; 37:109874. [PMID: 34686334 DOI: 10.1016/j.celrep.2021.109874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/02/2021] [Accepted: 09/30/2021] [Indexed: 12/18/2022] Open
Abstract
Embryos repair wounds rapidly, with no inflammation or scarring, in a process that involves polarization of the actomyosin cytoskeleton. Actomyosin polarization results in the assembly of a contractile cable around the wound that drives wound closure. Here, we demonstrate that a contractile actomyosin cable is not sufficient for rapid wound repair in Drosophila embryos. We show that wounding causes activation of the serine/threonine kinase p38 mitogen-activated protein kinase (MAPK) in the cells adjacent to the wound. p38 activation reduces the levels of wound-induced reactive oxygen species in the cells around the wound, limiting wound size. In addition, p38 promotes an increase in volume in the cells around the wound, thus facilitating the collective cell movements that drive rapid wound healing. Our data indicate that p38 regulates cell volumes through the sodium-potassium-chloride cotransporter NKCC1. Our work reveals cell growth and cell survival as cell behaviors critical for embryonic wound repair.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Miranda Victoria Hunter
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Ran Kafri
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
31
|
O’Connor J, Akbar FB, Hutson MS, Page-McCaw A. Zones of cellular damage around pulsed-laser wounds. PLoS One 2021; 16:e0253032. [PMID: 34570791 PMCID: PMC8476025 DOI: 10.1371/journal.pone.0253032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/07/2021] [Indexed: 11/19/2022] Open
Abstract
After a tissue is wounded, cells surrounding the wound adopt distinct wound-healing behaviors to repair the tissue. Considerable effort has been spent on understanding the signaling pathways that regulate immune and tissue-resident cells as they respond to wounds, but these signals must ultimately originate from the physical damage inflicted by the wound. Tissue wounds comprise several types of cellular damage, and recent work indicates that different types of cellular damage initiate different types of signaling. Hence to understand wound signaling, it is important to identify and localize the types of wound-induced cellular damage. Laser ablation is widely used by researchers to create reproducible, aseptic wounds in a tissue that can be live-imaged. Because laser wounding involves a combination of photochemical, photothermal and photomechanical mechanisms, each with distinct spatial dependencies, cells around a pulsed-laser wound will experience a gradient of damage. Here we exploit this gradient to create a map of wound-induced cellular damage. Using genetically-encoded fluorescent proteins, we monitor damaged cellular and sub-cellular components of epithelial cells in living Drosophila pupae in the seconds to minutes following wounding. We hypothesized that the regions of damage would be predictably arrayed around wounds of varying sizes, and subsequent analysis found that all damage radii are linearly related over a 3-fold range of wound size. Thus, around laser wounds, the distinct regions of damage can be estimated after measuring any one. This report identifies several different types of cellular damage within a wounded epithelial tissue in a living animal. By quantitatively mapping the size and placement of these different types of damage, we set the foundation for tracing wound-induced signaling back to the damage that initiates it.
Collapse
Affiliation(s)
- James O’Connor
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Fabiha Bushra Akbar
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - M. Shane Hutson
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
32
|
Sinenko SA, Starkova TY, Kuzmin AA, Tomilin AN. Physiological Signaling Functions of Reactive Oxygen Species in Stem Cells: From Flies to Man. Front Cell Dev Biol 2021; 9:714370. [PMID: 34422833 PMCID: PMC8377544 DOI: 10.3389/fcell.2021.714370] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS), superoxide anion and hydrogen peroxide, are generated as byproducts of oxidative phosphorylation in the mitochondria or via cell signaling-induced NADPH oxidases in the cytosol. In the recent two decades, a plethora of studies established that elevated ROS levels generated by oxidative eustress are crucial physiological mediators of many cellular and developmental processes. In this review, we discuss the mechanisms of ROS generation and regulation, current understanding of ROS functions in the maintenance of adult and embryonic stem cells, as well as in the process of cell reprogramming to a pluripotent state. Recently discovered cell-non-autonomous ROS functions mediated by growth factors are crucial for controlling cell differentiation and cellular immune response in Drosophila. Importantly, many physiological functions of ROS discovered in Drosophila may allow for deciphering and understanding analogous processes in human, which could potentially lead to the development of novel therapeutic approaches in ROS-associated diseases treatment.
Collapse
Affiliation(s)
- Sergey A Sinenko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Andrey A Kuzmin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexey N Tomilin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
33
|
Dunsmore G, Rosero EP, Shahbaz S, Santer DM, Jovel J, Lacy P, Houston S, Elahi S. Neutrophils promote T-cell activation through the regulated release of CD44-bound Galectin-9 from the cell surface during HIV infection. PLoS Biol 2021; 19:e3001387. [PMID: 34411088 PMCID: PMC8407585 DOI: 10.1371/journal.pbio.3001387] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/31/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
The interaction of neutrophils with T cells has been the subject of debate and controversies. Previous studies have suggested that neutrophils may suppress or activate T cells. Despite these studies, the interaction between neutrophils and T cells has remained a largely unexplored field. Here, based on our RNA sequencing (RNA-seq) analysis, we found that neutrophils have differential transcriptional and functional profiling depending on the CD4 T-cell count of the HIV-infected individual. In particular, we identified that neutrophils in healthy individuals express surface Galectin-9 (Gal-9), which is down-regulated upon activation, and is consistently down-regulated in HIV-infected individuals. However, down-regulation of Gal-9 was associated with CD4 T-cell count of patients. Unstimulated neutrophils express high levels of surface Gal-9 that is bound to CD44, and, upon stimulation, neutrophils depalmitoylate CD44 and induce its movement out of the lipid raft. This process causes the release of Gal-9 from the surface of neutrophils. In addition, we found that neutrophil-derived exogenous Gal-9 binds to cell surface CD44 on T cells, which promotes LCK activation and subsequently enhances T-cell activation. Furthermore, this process was regulated by glycolysis and can be inhibited by interleukin (IL)-10. Together, our data reveal a novel mechanism of Gal-9 shedding from the surface of neutrophils. This could explain elevated plasma Gal-9 levels in HIV-infected individuals as an underlying mechanism of the well-characterized chronic immune activation in HIV infection. This study provides a novel role for the Gal-9 shedding from neutrophils. We anticipate that our results will spark renewed investigation into the role of neutrophils in T-cell activation in other acute and chronic conditions, as well as improved strategies for modulating Gal-9 shedding. This study shows that HIV-infected individuals have different neutrophil profiles depending on their CD4 T cell count. In particular, neutrophils express high levels of surface Gal-9 but this is shed upon stimulation; this exogenous Gal-9 binds to CD44 on T cells, which promotes LCK activation and subsequently enhances T cell activation.
Collapse
Affiliation(s)
- Garett Dunsmore
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Eliana Perez Rosero
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shima Shahbaz
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Deanna M. Santer
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Juan Jovel
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Paige Lacy
- Department of Medicine, Division of Pulmonary Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Stan Houston
- Department of Medicine, Division of Infectious Disease, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shokrollah Elahi
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- * E-mail:
| |
Collapse
|
34
|
Gómez-Gálvez P, Anbari S, Escudero LM, Buceta J. Mechanics and self-organization in tissue development. Semin Cell Dev Biol 2021; 120:147-159. [PMID: 34417092 DOI: 10.1016/j.semcdb.2021.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023]
Abstract
Self-organization is an all-important feature of living systems that provides the means to achieve specialization and functionality at distinct spatio-temporal scales. Herein, we review this concept by addressing the packing organization of cells, the sorting/compartmentalization phenomenon of cell populations, and the propagation of organizing cues at the tissue level through traveling waves. We elaborate on how different theoretical models and tools from Topology, Physics, and Dynamical Systems have improved the understanding of self-organization by shedding light on the role played by mechanics as a driver of morphogenesis. Altogether, by providing a historical perspective, we show how ideas and hypotheses in the field have been revisited, developed, and/or rejected and what are the open questions that need to be tackled by future research.
Collapse
Affiliation(s)
- Pedro Gómez-Gálvez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla and Departamento de Biologia Celular, Universidad de Sevilla, 41013 Seville, Spain; Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Samira Anbari
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Luis M Escudero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla and Departamento de Biologia Celular, Universidad de Sevilla, 41013 Seville, Spain; Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Javier Buceta
- Institute for Integrative Systems Biology (I2SysBio), CSIC-UV, Paterna, 46980 Valencia, Spain.
| |
Collapse
|
35
|
Splitting up to heal: mitochondrial shape regulates signaling for focal membrane repair. Biochem Soc Trans 2021; 48:1995-2002. [PMID: 32985660 DOI: 10.1042/bst20200120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022]
Abstract
Mitochondria are central to the health of eukaryotic cells. While commonly known for their bioenergetic role, mitochondria also function as signaling organelles that regulate cell stress responses capable of restoring homeostasis or leading the stressed cell to eventual death. Damage to the plasma membrane is a potentially fatal stressor incurred by all cells. Repairing plasma membrane damage requires cells to mount a rapid and localized response to injury. Accumulating evidence has identified a role for mitochondria as an important facilitator of this acute and localized repair response. However, as mitochondria are organized in a cell-wide, interconnected network, it is unclear how they collectively sense and respond to a focal injury. Here we will discuss how mitochondrial shape change is an integral part of this localized repair response. Mitochondrial fragmentation spatially restricts beneficial repair signaling, enabling a localized response to focal injury. Conservation of mitochondrial fragmentation in response to cell and tissue damage across species demonstrates that this is a universal pro-survival adaptation to injury and suggests that mitochondrial fragmentation may provide cells a mechanism to facilitate localized signaling in contexts beyond repairing plasma membrane injury.
Collapse
|
36
|
O'Connor JT, Stevens AC, Shannon EK, Akbar FB, LaFever KS, Narayanan NP, Gailey CD, Hutson MS, Page-McCaw A. Proteolytic activation of Growth-blocking peptides triggers calcium responses through the GPCR Mthl10 during epithelial wound detection. Dev Cell 2021; 56:2160-2175.e5. [PMID: 34273275 DOI: 10.1016/j.devcel.2021.06.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/20/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022]
Abstract
The presence of a wound triggers surrounding cells to initiate repair mechanisms, but it is not clear how cells initially detect wounds. In epithelial cells, the earliest known wound response, occurring within seconds, is a dramatic increase in cytosolic calcium. Here, we show that wounds in the Drosophila notum trigger cytoplasmic calcium increase by activating extracellular cytokines, Growth-blocking peptides (Gbps), which initiate signaling in surrounding epithelial cells through the G-protein-coupled receptor Methuselah-like 10 (Mthl10). Latent Gbps are present in unwounded tissue and are activated by proteolytic cleavage. Using wing discs, we show that multiple protease families can activate Gbps, suggesting that they act as a generalized protease-detector system. We present experimental and computational evidence that proteases released during wound-induced cell damage and lysis serve as the instructive signal: these proteases liberate Gbp ligands, which bind to Mthl10 receptors on surrounding epithelial cells, and activate downstream release of calcium.
Collapse
Affiliation(s)
- James T O'Connor
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Aaron C Stevens
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, USA
| | - Erica K Shannon
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Fabiha Bushra Akbar
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Kimberly S LaFever
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Neil P Narayanan
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Casey D Gailey
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - M Shane Hutson
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA; Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA.
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
37
|
Hebbar S, Knust E. Reactive oxygen species (ROS) constitute an additional player in regulating epithelial development. Bioessays 2021; 43:e2100096. [PMID: 34260754 DOI: 10.1002/bies.202100096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules produced in cells. So far, they have mostly been connected to diseases and pathological conditions. More recent results revealed a somewhat unexpected role of ROS in control of developmental processes. In this review, we elaborate on ROS in development, focussing on their connection to epithelial tissue morphogenesis. After briefly summarising unique characteristics of epithelial cells, we present some characteristic features of ROS species, their production and targets, with a focus on proteins important for epithelial development and function. Finally, we provide examples of regulation of epithelial morphogenesis by ROS, and also of developmental genes that regulate the overall redox status. We conclude by discussing future avenues of research that will further elucidate ROS regulation in epithelial development.
Collapse
Affiliation(s)
- Sarita Hebbar
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
38
|
Dias Gomes M, Iden S. Orchestration of tissue-scale mechanics and fate decisions by polarity signalling. EMBO J 2021; 40:e106787. [PMID: 33998017 PMCID: PMC8204866 DOI: 10.15252/embj.2020106787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Eukaryotic development relies on dynamic cell shape changes and segregation of fate determinants to achieve coordinated compartmentalization at larger scale. Studies in invertebrates have identified polarity programmes essential for morphogenesis; however, less is known about their contribution to adult tissue maintenance. While polarity-dependent fate decisions in mammals utilize molecular machineries similar to invertebrates, the hierarchies and effectors can differ widely. Recent studies in epithelial systems disclosed an intriguing interplay of polarity proteins, adhesion molecules and mechanochemical pathways in tissue organization. Based on major advances in biophysics, genome editing, high-resolution imaging and mathematical modelling, the cell polarity field has evolved to a remarkably multidisciplinary ground. Here, we review emerging concepts how polarity and cell fate are coupled, with emphasis on tissue-scale mechanisms, mechanobiology and mammalian models. Recent findings on the role of polarity signalling for tissue mechanics, micro-environmental functions and fate choices in health and disease will be summarized.
Collapse
Affiliation(s)
- Martim Dias Gomes
- CECAD Cluster of ExcellenceUniversity of CologneCologneGermany
- Cell and Developmental BiologyFaculty of MedicineCenter of Human and Molecular Biology (ZHMB)Saarland UniversityHomburgGermany
| | - Sandra Iden
- CECAD Cluster of ExcellenceUniversity of CologneCologneGermany
- Cell and Developmental BiologyFaculty of MedicineCenter of Human and Molecular Biology (ZHMB)Saarland UniversityHomburgGermany
- CMMCUniversity of CologneCologneGermany
| |
Collapse
|
39
|
Tamada M, Shi J, Bourdot KS, Supriyatno S, Palmquist KH, Gutierrez-Ruiz OL, Zallen JA. Toll receptors remodel epithelia by directing planar-polarized Src and PI3K activity. Dev Cell 2021; 56:1589-1602.e9. [PMID: 33932332 DOI: 10.1016/j.devcel.2021.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/11/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Toll-like receptors are essential for animal development and survival, with conserved roles in innate immunity, tissue patterning, and cell behavior. The mechanisms by which Toll receptors signal to the nucleus are well characterized, but how Toll receptors generate rapid, localized signals at the cell membrane to produce acute changes in cell polarity and behavior is not known. We show that Drosophila Toll receptors direct epithelial convergent extension by inducing planar-polarized patterns of Src and PI3-kinase (PI3K) activity. Toll receptors target Src activity to specific sites at the membrane, and Src recruits PI3K to the Toll-2 complex through tyrosine phosphorylation of the Toll-2 cytoplasmic domain. Reducing Src or PI3K activity disrupts planar-polarized myosin assembly, cell intercalation, and convergent extension, whereas constitutive Src activity promotes ectopic PI3K and myosin cortical localization. These results demonstrate that Toll receptors direct cell polarity and behavior by locally mobilizing Src and PI3K activity.
Collapse
Affiliation(s)
- Masako Tamada
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Jay Shi
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Kia S Bourdot
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Sara Supriyatno
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Karl H Palmquist
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Omar L Gutierrez-Ruiz
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
40
|
Dwivedi S, Kushalan S, Paithankar JG, D'Souza LC, Hegde S, Sharma A. Environmental toxicants, oxidative stress and health adversities: interventions of phytochemicals. J Pharm Pharmacol 2021; 74:516-536. [PMID: 33822130 DOI: 10.1093/jpp/rgab044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Oxidative stress is the most common factor mediating environmental chemical-induced health adversities. Recently, an exponential rise in the use of phytochemicals as an alternative therapeutics against oxidative stress-mediated diseases has been documented. Due to their free radical quenching property, plant-derived natural products have gained substantial attention as a therapeutic agent in environmental toxicology. The present review aimed to describe the therapeutic role of phytochemicals in mitigating environmental toxicant-mediated sub-cellular and organ toxicities via controlling cellular antioxidant response. METHODS The present review has covered the recently related studies, mainly focussing on the free radical scavenging role of phytochemicals in environmental toxicology. KEY FINDINGS In vitro and in vivo studies have reported that supplementation of antioxidant-rich compounds can ameliorate the toxicant-induced oxidative stress, thereby improving the health conditions. Improving the cellular antioxidant pool has been considered as a mode of action of phytochemicals. However, the other cellular targets of phytochemicals remain uncertain. CONCLUSIONS Knowing the therapeutic value of phytochemicals to mitigate the chemical-induced toxicity is an initial stage; mechanistic understanding needs to decipher for development as therapeutics. Moreover, examining the efficacy of phytochemicals against mixer toxicity and identifying the bioactive molecule are major challenges in the field.
Collapse
Affiliation(s)
- Shiwangi Dwivedi
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Sharanya Kushalan
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Jagdish Gopal Paithankar
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Smitha Hegde
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| |
Collapse
|
41
|
Garcez M, Branco-Santos J, Gracio PC, Homem CCF. Mitochondrial Dynamics in the Drosophila Ovary Regulates Germ Stem Cell Number, Cell Fate, and Female Fertility. Front Cell Dev Biol 2021; 8:596819. [PMID: 33585443 PMCID: PMC7876242 DOI: 10.3389/fcell.2020.596819] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/30/2020] [Indexed: 01/07/2023] Open
Abstract
The fate and proliferative capacity of stem cells have been shown to strongly depend on their metabolic state. Mitochondria are the powerhouses of the cell being responsible for energy production via oxidative phosphorylation (OxPhos) as well as for several other metabolic pathways. Mitochondrial activity strongly depends on their structural organization, with their size and shape being regulated by mitochondrial fusion and fission, a process known as mitochondrial dynamics. However, the significance of mitochondrial dynamics in the regulation of stem cell metabolism and fate remains elusive. Here, we characterize the role of mitochondria morphology in female germ stem cells (GSCs) and in their more differentiated lineage. Mitochondria are particularly important in the female GSC lineage. Not only do they provide these cells with their energy requirements to generate the oocyte but they are also the only mitochondria pool to be inherited by the offspring. We show that the undifferentiated GSCs predominantly have fissed mitochondria, whereas more differentiated germ cells have more fused mitochondria. By reducing the levels of mitochondrial dynamics regulators, we show that both fused and fissed mitochondria are required for the maintenance of a stable GSC pool. Surprisingly, we found that disrupting mitochondrial dynamics in the germline also strongly affects nurse cells morphology, impairing egg chamber development and female fertility. Interestingly, reducing the levels of key enzymes in the Tricarboxylic Acid Cycle (TCA), known to cause OxPhos reduction, also affects GSC number. This defect in GSC self-renewal capacity indicates that at least basal levels of TCA/OxPhos are required in GSCs. Our findings show that mitochondrial dynamics is essential for female GSC maintenance and female fertility, and that mitochondria fusion and fission events are dynamically regulated during GSC differentiation, possibly to modulate their metabolic profile.
Collapse
Affiliation(s)
- Marcia Garcez
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Universidade do Porto, Porto, Portugal
| | - Joana Branco-Santos
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Patricia C Gracio
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Catarina C F Homem
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
42
|
Scepanovic G, Florea A, Fernandez-Gonzalez R. Multiscale In Vivo Imaging of Collective Cell Migration in Drosophila Embryos. Methods Mol Biol 2021; 2179:199-224. [PMID: 32939723 DOI: 10.1007/978-1-0716-0779-4_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Coordinated cell movements drive embryonic development and tissue repair, and can also spread disease. Time-lapse microscopy is an integral part in the study of the cell biology of collective cell movements. Advances in imaging techniques enable monitoring dynamic cellular and molecular events in real time within living animals. Here, we demonstrate the use of spinning disk confocal microscopy to investigate coordinated cell movements and epithelial-to-mesenchymal-like transitions during embryonic wound closure in Drosophila. We describe image-based metrics to quantify the efficiency of collective cell migration. Finally, we show the application of super-resolution radial fluctuation microscopy to obtain multidimensional, super-resolution images of protrusive activity in collectively moving cells in vivo. Together, the methods presented here constitute a toolkit for the modern analysis of collective cell migration in living animals.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Alexandru Florea
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada. .,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada. .,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada. .,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
43
|
Chakrabarti S, Visweswariah SS. Intramacrophage ROS Primes the Innate Immune System via JAK/STAT and Toll Activation. Cell Rep 2020; 33:108368. [PMID: 33176146 PMCID: PMC7662148 DOI: 10.1016/j.celrep.2020.108368] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/10/2019] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Tissue injury is one of the most severe environmental perturbations for a living organism. When damage occurs in adult Drosophila, there is a local response of the injured tissue and a coordinated action across different tissues to help the organism overcome the deleterious effect of an injury. We show a change in the transcriptome of hemocytes at the site of tissue injury, with pronounced activation of the Toll signaling pathway. We find that induction of the cytokine upd-3 and Toll receptor activation occur in response to injury alone, in the absence of a pathogen. Intracellular accumulation of hydrogen peroxide in hemocytes is essential for upd-3 induction and is facilitated by the diffusion of hydrogen peroxide through a channel protein Prip. Importantly, hemocyte activation and production of reactive oxygen species (ROS) at the site of a sterile injury provide protection to flies on subsequent infection, demonstrating training of the innate immune system.
Collapse
Affiliation(s)
- Sveta Chakrabarti
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India.
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
44
|
Teo JL, Tomatis VM, Coburn L, Lagendijk AK, Schouwenaar IM, Budnar S, Hall TE, Verma S, McLachlan RW, Hogan BM, Parton RG, Yap AS, Gomez GA. Src kinases relax adherens junctions between the neighbors of apoptotic cells to permit apical extrusion. Mol Biol Cell 2020; 31:2557-2569. [PMID: 32903148 PMCID: PMC7851871 DOI: 10.1091/mbc.e20-01-0084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 08/12/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelia can eliminate apoptotic cells by apical extrusion. This is a complex morphogenetic event where expulsion of the apoptotic cell is accompanied by rearrangement of its immediate neighbors to form a rosette. A key mechanism for extrusion is constriction of an actomyosin network that neighbor cells form at their interface with the apoptotic cell. Here we report a complementary process of cytoskeletal relaxation that occurs when cortical contractility is down-regulated at the junctions between those neighbor cells themselves. This reflects a mechanosensitive Src family kinase (SFK) signaling pathway that is activated in neighbor cells when the apoptotic cell relaxes shortly after injury. Inhibiting SFK signaling blocks both the expulsion of apoptotic cells and the rosette formation among their neighbor cells. This reveals the complex pattern of spatially distinct contraction and relaxation that must be established in the neighboring epithelium for apoptotic cells to be extruded.
Collapse
Affiliation(s)
- Jessica L. Teo
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Vanesa M. Tomatis
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Luke Coburn
- Institute of Complex Systems and Mathematical Biology, University of Aberdeen, Aberdeen, United Kingdom, AB24 3UE
| | - Anne K. Lagendijk
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Irin-Maya Schouwenaar
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Srikanth Budnar
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Thomas E. Hall
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Suzie Verma
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Robert W. McLachlan
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Benjamin M. Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Robert G. Parton
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
- Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Alpha S. Yap
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
| | - Guillermo A. Gomez
- Division of Cell and Developmental Biology, The University of Queensland, St Lucia, Queensland, Australia, 4072
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia, 5000
| |
Collapse
|
45
|
Breus O, Dickmeis T. Genetically encoded thiol redox-sensors in the zebrafish model: lessons for embryonic development and regeneration. Biol Chem 2020; 402:363-378. [PMID: 33021959 DOI: 10.1515/hsz-2020-0269] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Important roles for reactive oxygen species (ROS) and redox signaling in embryonic development and regenerative processes are increasingly recognized. However, it is difficult to obtain information on spatiotemporal dynamics of ROS production and signaling in vivo. The zebrafish is an excellent model for in vivo bioimaging and possesses a remarkable regenerative capacity upon tissue injury. Here, we review data obtained in this model system with genetically encoded redox-sensors targeting H2O2 and glutathione redox potential. We describe how such observations have prompted insight into regulation and downstream effects of redox alterations during tissue differentiation, morphogenesis and regeneration. We also discuss the properties of the different sensors and their consequences for the interpretation of in vivo imaging results. Finally, we highlight open questions and additional research fields that may benefit from further application of such sensor systems in zebrafish models of development, regeneration and disease.
Collapse
Affiliation(s)
- Oksana Breus
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| | - Thomas Dickmeis
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
46
|
Chowdhary S, Madan S, Tomer D, Mavrakis M, Rikhy R. Mitochondrial morphology and activity regulate furrow ingression and contractile ring dynamics in Drosophila cellularization. Mol Biol Cell 2020; 31:2331-2347. [PMID: 32755438 PMCID: PMC7851960 DOI: 10.1091/mbc.e20-03-0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mitochondria are maternally inherited in many organisms. Mitochondrial morphology and activity regulation is essential for cell survival, differentiation, and migration. An analysis of mitochondrial dynamics and function in morphogenetic events in early metazoan embryogenesis has not been carried out. In our study we find a crucial role of mitochondrial morphology regulation in cell formation in Drosophila embryogenesis. We find that mitochondria are small and fragmented and translocate apically on microtubules and distribute progressively along the cell length during cellularization. Embryos mutant for the mitochondrial fission protein, Drp1 (dynamin-related protein 1), die in embryogenesis and show an accumulation of clustered mitochondria on the basal side in cellularization. Additionally, Drp1 mutant embryos contain lower levels of reactive oxygen species (ROS). ROS depletion was previously shown to decrease myosin II activity. Drp1 loss also leads to myosin II depletion at the membrane furrow, thereby resulting in decreased cell height and larger contractile ring area in cellularization similar to that in myosin II mutants. The mitochondrial morphology and cellularization defects in Drp1 mutants are suppressed by reducing mitochondrial fusion and increasing cytoplasmic ROS in superoxide dismutase mutants. Our data show a key role for mitochondrial morphology and activity in supporting the morphogenetic events that drive cellularization in Drosophila embryos.
Collapse
Affiliation(s)
- Sayali Chowdhary
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Somya Madan
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Darshika Tomer
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Manos Mavrakis
- Aix Marseille University, CNRS, Centrale Marseille, Institut Fresnel, 13013 Marseille, France
| | - Richa Rikhy
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| |
Collapse
|
47
|
Benzarti M, Delbrouck C, Neises L, Kiweler N, Meiser J. Metabolic Potential of Cancer Cells in Context of the Metastatic Cascade. Cells 2020; 9:E2035. [PMID: 32899554 PMCID: PMC7563895 DOI: 10.3390/cells9092035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
The metastatic cascade is a highly plastic and dynamic process dominated by cellular heterogeneity and varying metabolic requirements. During this cascade, the three major metabolic pillars, namely biosynthesis, RedOx balance, and bioenergetics, have variable importance. Biosynthesis has superior significance during the proliferation-dominated steps of primary tumour growth and secondary macrometastasis formation and only minor relevance during the growth-independent processes of invasion and dissemination. Consequently, RedOx homeostasis and bioenergetics emerge as conceivable metabolic key determinants in cancer cells that disseminate from the primary tumour. Within this review, we summarise our current understanding on how cancer cells adjust their metabolism in the context of different microenvironments along the metastatic cascade. With the example of one-carbon metabolism, we establish a conceptual view on how the same metabolic pathway can be exploited in different ways depending on the current cellular needs during metastatic progression.
Collapse
Affiliation(s)
- Mohaned Benzarti
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Catherine Delbrouck
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Laura Neises
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Nicole Kiweler
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| |
Collapse
|
48
|
Armitage EL, Roddie HG, Evans IR. Overexposure to apoptosis via disrupted glial specification perturbs Drosophila macrophage function and reveals roles of the CNS during injury. Cell Death Dis 2020; 11:627. [PMID: 32796812 PMCID: PMC7428013 DOI: 10.1038/s41419-020-02875-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Apoptotic cell clearance by phagocytes is a fundamental process during development, homeostasis and the resolution of inflammation. However, the demands placed on phagocytic cells such as macrophages by this process, and the limitations these interactions impose on subsequent cellular behaviours are not yet clear. Here, we seek to understand how apoptotic cells affect macrophage function in the context of a genetically tractable Drosophila model in which macrophages encounter excessive amounts of apoptotic cells. Loss of the glial-specific transcription factor Repo prevents glia from contributing to apoptotic cell clearance in the developing embryo. We show that this leads to the challenge of macrophages with large numbers of apoptotic cells in vivo. As a consequence, macrophages become highly vacuolated with cleared apoptotic cells, and their developmental dispersal and migration is perturbed. We also show that the requirement to deal with excess apoptosis caused by a loss of repo function leads to impaired inflammatory responses to injury. However, in contrast to migratory phenotypes, defects in wound responses cannot be rescued by preventing apoptosis from occurring within a repo mutant background. In investigating the underlying cause of these impaired inflammatory responses, we demonstrate that wound-induced calcium waves propagate into surrounding tissues, including neurons and glia of the ventral nerve cord, which exhibit striking calcium waves on wounding, revealing a previously unanticipated contribution of these cells during responses to injury. Taken together, these results demonstrate important insights into macrophage biology and how repo mutants can be used to study macrophage-apoptotic cell interactions in the fly embryo. Furthermore, this work shows how these multipurpose cells can be 'overtasked' to the detriment of their other functions, alongside providing new insights into which cells govern macrophage responses to injury in vivo.
Collapse
Affiliation(s)
- Emma Louise Armitage
- Department of Infection, Immunity and Cardiovascular Disease and The Bateson Centre, University of Sheffield, Sheffield, UK
| | - Hannah Grace Roddie
- Department of Infection, Immunity and Cardiovascular Disease and The Bateson Centre, University of Sheffield, Sheffield, UK
| | - Iwan Robert Evans
- Department of Infection, Immunity and Cardiovascular Disease and The Bateson Centre, University of Sheffield, Sheffield, UK.
| |
Collapse
|
49
|
Biel N, Figard L, Sokac AM. Imaging Intranuclear Actin Rods in Live Heat Stressed Drosophila Embryos. J Vis Exp 2020. [PMID: 32478727 DOI: 10.3791/61297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The purpose of this protocol is to visualize intranuclear actin rods that assemble in live Drosophila melanogaster embryos following heat stress. Actin rods are a hallmark of a conserved, inducible Actin Stress Response (ASR) that accompanies human pathologies, including neurodegenerative disease. Previously, we showed that the ASR contributes to morphogenesis failures and reduced viability of developing embryos. This protocol allows the continued study of mechanisms underlying actin rod assembly and the ASR in a model system that is highly amenable to imaging, genetics and biochemistry. Embryos are collected and mounted on a coverslip to prepare them for injection. Rhodamine-conjugated globular actin (G-actinRed) is diluted and loaded into a microneedle. A single injection is made into the center of each embryo. After injection, embryos are incubated at elevated temperature and intranuclear actin rods are then visualized by confocal microscopy. Fluorescence recovery after photobleaching (FRAP) experiments may be performed on the actin rods; and other actin-rich structures in the cytoplasm can also be imaged. We find that G-actinRed polymerizes like endogenous G-actin and does not, on its own, interfere with normal embryo development. One limitation of this protocol is that care must be taken during injection to avoid serious injury to the embryo. However, with practice, injecting G-actinRed into Drosophila embryos is a fast and reliable way to visualize actin rods and can easily be used with flies of any genotype or with the introduction of other cellular stresses, including hypoxia and oxidative stress.
Collapse
Affiliation(s)
- Natalie Biel
- Integrative Molecular and Biomedical Sciences, Baylor College of Medicine; Department of Cell and Molecular Biology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign
| | - Lauren Figard
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine
| | - Anna Marie Sokac
- Integrative Molecular and Biomedical Sciences, Baylor College of Medicine; Department of Cell and Molecular Biology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine;
| |
Collapse
|
50
|
Ponte S, Carvalho L, Gagliardi M, Campos I, Oliveira PJ, Jacinto A. Drp1-mediated mitochondrial fission regulates calcium and F-actin dynamics during wound healing. Biol Open 2020; 9:bio048629. [PMID: 32184231 PMCID: PMC7225088 DOI: 10.1242/bio.048629] [Citation(s) in RCA: 20] [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: 10/07/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondria adapt to cellular needs by changes in morphology through fusion and fission events, referred to as mitochondrial dynamics. Mitochondrial function and morphology are intimately connected and the dysregulation of mitochondrial dynamics is linked to several human diseases. In this work, we investigated the role of mitochondrial dynamics in wound healing in the Drosophila embryonic epidermis. Mutants for mitochondrial fusion and fission proteins fail to close their wounds, indicating that the regulation of mitochondrial dynamics is required for wound healing. By live-imaging, we found that loss of function of the mitochondrial fission protein Dynamin-related protein 1 (Drp1) compromises the increase of cytosolic and mitochondrial calcium upon wounding and leads to reduced reactive oxygen species (ROS) production and F-actin defects at the wound edge, culminating in wound healing impairment. Our results highlight a new role for mitochondrial dynamics in the regulation of calcium, ROS and F-actin during epithelial repair.
Collapse
Affiliation(s)
- Susana Ponte
- CEDOC, Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Lara Carvalho
- CEDOC, Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Maria Gagliardi
- CEDOC, Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Isabel Campos
- Animal Platforms, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Paulo J Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, 3060-197 Cantanhede, Portugal
| | - António Jacinto
- CEDOC, Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| |
Collapse
|