Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Heliyon ; 10(4): e26323, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38404824

ABSTRACT

The integration of photovoltaic technologies within the agricultural framework, known as agrivoltaics, emerges as a promising and sustainable solution to meet the growing global demands for energy and food production. This innovative technology enables the simultaneous utilization of sunlight for both photovoltaics (PV) and photosynthesis. A key challenge in agrivoltaic research involves identifying technologies applicable to a wide range of plant species and diverse geographic regions. To address this challenge, we adopt a multi-experimental and multi-species approach to assess the viability of semi-transparent, spectrally selective thin-film silicon PV technology. Our findings demonstrate compatibility with crop production in controlled environments for both plants and algae. Notably, selective thin-film PV exhibits the potential to enhance crop yields and serves as a photo-protectant. We observe that plant and algal growth increases beneath the selective PV film when supplemented with appropriate diffuse light in the growth environment. Conversely, in situations where light intensity exceeds optimal levels for plant growth, the selective PV film provides a photo-protective effect. These results suggest potential supplementary benefits of employing this technology in regions characterized by excessive light irradiation, where it can contribute to healthy plant growth.

2.
Front Cell Dev Biol ; 8: 572094, 2020.
Article in English | MEDLINE | ID: mdl-33043004

ABSTRACT

At the end of abscission, the residual midbody forms the so-called midbody remnant (MBR), a platform affecting cell fate with emerging key role in differentiation, development, and tumorigenicity. Depending on cell type and pathophysiological context, MBRs undergo different outcomes: they can be retained, released, internalized by nearby cells, or removed through autophagy-mediated degradation. Although mechanisms underlying MBR formation, positioning, and processing have been recently identified, their regulation is still largely unknown. Here, we report that the multifunctional kinase HIPK2 regulates MBR processing contributing to MBR removal. In the process of studying the role of HIPK2 in abscission, we observed that, in addition to cytokinesis failure, HIPK2 depletion leads to significant accumulation of MBRs. In particular, we detected comparable accumulation of MBRs after HIPK2 depletion or treatment with the autophagic inhibitor chloroquine. In contrast, single depletion of the two independent HIPK2 abscission targets, extrachromosomal histone H2B and severing enzyme Spastin, only marginally increased MBR retention, suggesting that MBR accumulation is not just linked to cytokinesis failure. We found that HIPK2 depletion leads to (i) increased levels of CEP55, a key effector of both midbody formation and MBR degradation; (ii) decreased levels of the selective autophagy receptors NBR1 and p62/SQSTM1; and (iii) impaired autophagic flux. These data suggest that HIPK2 contributes to MBR processing by regulating its autophagy-mediated degradation.

3.
Life Sci Alliance ; 3(12)2020 12.
Article in English | MEDLINE | ID: mdl-33106322

ABSTRACT

Hereditary Spastic Paraplegia (HSP) is a neurodegenerative disease most commonly caused by autosomal dominant mutations in the SPG4 gene encoding the microtubule-severing protein spastin. We hypothesise that SPG4-HSP is attributable to reduced spastin function because of haploinsufficiency; thus, therapeutic approaches which elevate levels of the wild-type spastin allele may be an effective therapy. However, until now, how spastin levels are regulated is largely unknown. Here, we show that the kinase HIPK2 regulates spastin protein levels in proliferating cells, in differentiated neurons and in vivo. Our work reveals that HIPK2-mediated phosphorylation of spastin at S268 inhibits spastin K48-poly-ubiquitination at K554 and prevents its neddylation-dependent proteasomal degradation. In a spastin RNAi neuronal cell model, overexpression of HIPK2, or inhibition of neddylation, restores spastin levels and rescues neurite defects. Notably, we demonstrate that spastin levels can be restored pharmacologically by inhibiting its neddylation-mediated degradation in neurons derived from a spastin mouse model of HSP and in patient-derived cells, thus revealing novel therapeutic targets for the treatment of SPG4-HSP.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Spastic Paraplegia, Hereditary/metabolism , Spastin/metabolism , Animals , Carrier Proteins/physiology , Disease Models, Animal , Gene Expression Regulation/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Microtubules/metabolism , Mutation , Neurites/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Protein Serine-Threonine Kinases/physiology , Proteolysis , Spastic Paraplegia, Hereditary/physiopathology , Spastin/physiology , Synapses/metabolism , Ubiquitination
4.
Cells ; 9(2)2020 02 20.
Article in English | MEDLINE | ID: mdl-32093146

ABSTRACT

HIPK2 is a DYRK-like kinase involved in cellular stress response pathways, development, and cell division. Two alternative splice variants of HIPK2, HIPK2-FL and HIPK2-Δe8, have been previously identified as having different protein stability but similar functional activity in the stress response. Here, we describe one additional HIPK2 splice variant with a distinct subcellular distribution and functional activity in cytokinesis. This novel splice variant lacks the last two exons and retains intron13 with a stop codon after 89 bp of the intron, generating a short isoform, HIPK2-S, that is detectable by 2D Western blots. RT-PCR analyses of tissue arrays and tumor samples show that HIPK2-FL and HIPK2-S are expressed in normal human tissues in a tissue-dependent manner and differentially expressed in human colorectal and pancreatic cancers. Gain- and loss-of-function experiments showed that in contrast to HIPK2-FL, HIPK2-S has a diffuse, non-speckled distribution and is not involved in the DNA damage response. Rather, we found that HIPK2-S, but not HIPK2-FL, localizes at the intercellular bridge, where it phosphorylates histone H2B and spastin, both required for faithful cell division. Altogether, these data show that distinct human HIPK2 splice variants are involved in distinct HIPK2-regulated functions like stress response and cytokinesis.


Subject(s)
Alternative Splicing/genetics , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cytokinesis/genetics , Introns , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Carrier Proteins/genetics , Codon, Terminator , Exons , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Spastin/metabolism , Transfection
5.
Cells ; 8(7)2019 07 05.
Article in English | MEDLINE | ID: mdl-31284535

ABSTRACT

Abscission is the final step of cell division, mediating the physical separation of the two daughter cells. A key player in this process is the microtubule-severing enzyme spastin that localizes at the midbody where its activity is crucial to cut microtubules and culminate the cytokinesis. Recently, we demonstrated that HIPK2, a multifunctional kinase involved in several cellular pathways, contributes to abscission and prevents tetraploidization. Here, we show that HIPK2 binds and phosphorylates spastin at serine 268. During cytokinesis, the midbody-localized spastin is phosphorylated at S268 in HIPK2-proficient cells. In contrast, no spastin is detectable at the midbody in HIPK2-depleted cells. The non-phosphorylatable spastin-S268A mutant does not localize at the midbody and cannot rescue HIPK2-depleted cells from abscission defects. In contrast, the phosphomimetic spastin-S268D mutant localizes at the midbody and restores successful abscission in the HIPK2-depleted cells. These results show that spastin is a novel target of HIPK2 and that HIPK2-mediated phosphorylation of spastin contributes to its midbody localization for successful abscission.


Subject(s)
Carrier Proteins/metabolism , Cytokinesis , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Spastin/metabolism , Cell Line, Tumor , Humans , Mutagenesis, Site-Directed , Phosphorylation , Serine/genetics , Serine/metabolism , Spastin/genetics
6.
Oncogene ; 37(26): 3562-3574, 2018 06.
Article in English | MEDLINE | ID: mdl-29563611

ABSTRACT

Cytokinesis, the final phase of cell division, is necessary to form two distinct daughter cells with correct distribution of genomic and cytoplasmic materials. Its failure provokes genetically unstable states, such as tetraploidization and polyploidization, which can contribute to tumorigenesis. Aurora-B kinase controls multiple cytokinetic events, from chromosome condensation to abscission when the midbody is severed. We have previously shown that HIPK2, a kinase involved in DNA damage response and development, localizes at the midbody and contributes to abscission by phosphorylating extrachromosomal histone H2B at Ser14. Of relevance, HIPK2-defective cells do not phosphorylate H2B and do not successfully complete cytokinesis leading to accumulation of binucleated cells, chromosomal instability, and increased tumorigenicity. However, how HIPK2 and H2B are recruited to the midbody during cytokinesis is still unknown. Here, we show that regardless of their direct (H2B) and indirect (HIPK2) binding of chromosomal DNA, both H2B and HIPK2 localize at the midbody independently of nucleic acids. Instead, by using mitotic kinase-specific inhibitors in a spatio-temporal regulated manner, we found that Aurora-B kinase activity is required to recruit both HIPK2 and H2B to the midbody. Molecular characterization showed that Aurora-B directly binds and phosphorylates H2B at Ser32 while indirectly recruits HIPK2 through the central spindle components MgcRacGAP and PRC1. Thus, among different cytokinetic functions, Aurora-B separately recruits HIPK2 and H2B to the midbody and these activities contribute to faithful cytokinesis.


Subject(s)
Aurora Kinase B/metabolism , Carrier Proteins/metabolism , Cytokinesis/physiology , Histones/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosomal Instability/genetics , DNA-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , RNA Interference , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL