Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Exp Cell Res ; 352(1): 95-103, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28161539

ABSTRACT

KIF3b is a protein of the kinesin-2 family which plays an important role in intraflagellar transport. Testis cancer is a common cancer among young men. Its diagnostic rate is increasing and over half of the cases are seminomas. Many aspects of the mechanism and gene expression background of this cancer remain unclear. Using western-blotting and semi-quantitative PCR we found high protein levels of KIF3b enrichment in seminoma tissue despite the mRNA levels remaining equivalent to that of normal testicular tissues. The distribution of KIF3b was mainly in cells with division potential. Wound-healing assays and cell counting kit assays showed that the knockdown of KIF3b significantly suppressed cell migration ability, viability and number in HeLa cells. Immunofluorescence images during the cell cycle revealed that KIF3b tended to gather at the spindles and was enriched at the central spindle. This indicated that KIF3b may also have direct impacts upon spindle formation and cytokinesis. By counting the numbers of nuclei, spindles and cells, we found that the rates of multipolar division and multi-nucleation were raised in KIF3b-knockdown cells. In this way we demonstrate that KIF3b functions importantly in mitosis and may be essential to seminoma cell division and proliferation as well as being necessary for normal cell division.


Subject(s)
Cell Cycle , Cytokinesis/physiology , Kinesins/metabolism , Mitosis/physiology , Seminoma/pathology , Testicular Neoplasms/pathology , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Fluorescent Antibody Technique , HeLa Cells , Humans , Kinesins/genetics , Male , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Seminoma/genetics , Seminoma/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Tumor Cells, Cultured
2.
Cell Rep ; 43(7): 114417, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38980795

ABSTRACT

The ability to sense and respond to osmotic fluctuations is critical for the maintenance of cellular integrity. We used gene co-essentiality analysis to identify an unappreciated relationship between TSC22D2, WNK1, and NRBP1 in regulating cell volume homeostasis. All of these genes have paralogs and are functionally buffered for osmo-sensing and cell volume control. Within seconds of hyperosmotic stress, TSC22D, WNK, and NRBP family members physically associate into biomolecular condensates, a process that is dependent on intrinsically disordered regions (IDRs). A close examination of these protein families across metazoans revealed that TSC22D genes evolved alongside a domain in NRBPs that specifically binds to TSC22D proteins, which we have termed NbrT (NRBP binding region with TSC22D), and this co-evolution is accompanied by rapid IDR length expansion in WNK-family kinases. Our study reveals that TSC22D, WNK, and NRBP genes evolved in metazoans to co-regulate rapid cell volume changes in response to osmolarity.


Subject(s)
Cell Size , WNK Lysine-Deficient Protein Kinase 1 , Humans , Animals , WNK Lysine-Deficient Protein Kinase 1/metabolism , WNK Lysine-Deficient Protein Kinase 1/genetics , Evolution, Molecular , HEK293 Cells , Protein Binding , Multigene Family , Osmotic Pressure
3.
Oncotarget ; 8(37): 61373-61384, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28977870

ABSTRACT

C-terminus kinesin motor KIFC1 is known for centrosome clustering in cancer cells with supernumerary centrosomes. KIFC1 crosslinks and glides on microtubules (MT) to assist normal bipolar spindle formation to avoid multi-polar cell division, which might be fatal. Testis cancer is the most common human cancer among young men. However, the gene expression profiles of testis cancer is still not complete and the expression of the C-terminus kinesin motor KIFC1 in testis cancer has not yet been examined. We found that KIFC1 is enriched in seminoma tissues in both mRNA level and protein level, and is specifically enriched in the cells that divide actively. Cell experiments showed that KIFC1 may be essential in cell division, but not essential in metastasis. Based on subcellular immuno-florescent staining results, we also described the localization of KIFC1 during cell cycle. By expressing ΔC-FLAG peptide in the cells, we found that the tail domain of KIFC1 might be essential for the dynamic disassociation of KIFC1, and the motor domain of KIFC1 might be essential for the degradation of KIFC1. Our work provides a new perspective for seminoma research.

4.
Oncotarget ; 7(30): 48656-48670, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27102297

ABSTRACT

The kinesin motor KIFC1 has been suggested as a potential chemotherapy target due to its critical role in clustering of the multiple centrosomes found in cancer cells. In this regard, KIFC1 seems to be non-essential in normal somatic cells which usually possess only two centrosomes. Moreover, KIFC1 is also found to initiatively drive tumor malignancy and metastasis by stabilizing a certain degree of genetic instability, delaying cell cycle and protecting cancer cell surviving signals. However, that KIFC1 also plays roles in other specific cell types complicates the question of whether it is a promising chemotherapy target for cancer treatment. For example, KIFC1 is found functionally significant in vesicular and organelle trafficking, spermiogenesis, oocyte development, embryo gestation and double-strand DNA transportation. In this review we summarize a recent collection of information so as to provide a generalized picture of ideas and mechanisms against and in favor of KIFC1 as a chemotherapy target. And we also drew the conclusion that KIFC1 is a promising chemotherapy target for some types of cancers, because the side-effects of inhibiting KIFC1 mentioned in this review are theoretically easy to avoid, while KIFC1 is functionally indispensable during mitosis and malignancy of multi-centrosome cancer cells. Further investigations of how KIFC1 is regulated throughout the mitosis in cancer cells are needed for the understanding of the pathways where KIFC1 is involved and for further exploitation of indirect KIFC1 inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Centrosome/drug effects , Kinesins/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Alanine/analogs & derivatives , Alanine/therapeutic use , Centrosome/metabolism , DNA/drug effects , DNA/metabolism , Female , Humans , Kinesins/metabolism , Mitosis , Molecular Targeted Therapy/adverse effects , Neoplasms/pathology , Oocytes/drug effects , Oocytes/growth & development , Phenanthrenes/therapeutic use , Phenylalanine/analogs & derivatives , Phenylalanine/therapeutic use , Pregnancy/drug effects , Protein Transport/drug effects , Pyridines/therapeutic use , Signal Transduction/drug effects , Spermatogenesis/drug effects , ortho-Aminobenzoates/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL