Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rabia Islam is active.

Publication


Featured researches published by Rabia Islam.


Journal of Biological Chemistry | 2014

Prolyl isomerase Pin1-mediated conformational change and subnuclear focal accumulation of Runx2 are crucial for fibroblast growth factor 2 (FGF2)-induced osteoblast differentiation.

Won-Joon Yoon; Young-Dan Cho; Woo-Jin Kim; Han-Sol Bae; Rabia Islam; Kyung Mi Woo; Jeong-Hwa Baek; Suk-Chul Bae; Hyun-Mo Ryoo

Background: Genetic interaction between Runx2 and Pin1 is critical for embryonic bone formation. Results: Pin1 is a critical modifying enzyme promoting both subnuclear accumulation and protein acetylation of Runx2. Conclusion: Pin1 determines the fate of Runx2 protein in osteoblast differentiation. Significance: The modulation of Pin1 activity may be a clinical target for the regulation of bone formation. Fibroblast growth factor 2 (FGF2) signaling plays a pivotal role in bone growth/differentiation through the activation of osteogenic master transcription factor Runx2, which is mediated by the ERK/MAPK-dependent phosphorylation and the p300-dependent acetylation of Runx2. In this study, we found that Pin1-dependent isomerization of Runx2 is the critical step for FGF2-induced Runx2 transactivation function. We identified four serine or threonine residues in the C-terminal domain of Runx2 that are responsible for Pin1 binding and structural modification. Confocal imaging studies indicated that FGF2 treatment strongly stimulated the focal accumulation of Pin1 in the subnuclear area, which recruited Runx2. In addition, active forms of RNA polymerase-II also colocalized in the same subnuclear compartment. Dipentamethylene thiuram monosulfide, a Pin1 inhibitor, strongly attenuated their focal accumulation as well as Runx2 transactivation activity. The Pin1-mediated structural modification of Runx2 is an indispensable step connecting phosphorylation and acetylation and, consequently, transcriptional activation of Runx2 by FGF signaling. Thus, the modulation of Pin1 activity may be a target for the regulation of bone formation.


Journal of Cellular Physiology | 2014

Pin1 regulates osteoclast fusion through suppression of the master regulator of cell fusion DC-STAMP.

Rabia Islam; Han-Sol Bae; Won-Joon Yoon; Kyung Mi Woo; Jeong-Hwa Baek; Hong‐Hee Kim; Takafumi Uchida; Hyun-Mo Ryoo

Cell fusion is a fundamental biological event that is essential for the development of multinucleated cells such as osteoclasts. Fusion failure leads to the accumulation of dense bone such as in osteopetrosis, demonstrating the importance of fusion in osteoclast maturity and bone remodeling. In a recent study, we reported that Pin1 plays a role in the regulation of bone formation and Runx2 regulation. In this study, we explored the role of Pin1 in osteoclast formation and bone resorption. Pin1 null mice have low bone mass and increased TRAP staining in histological sections of long bones, compared to Pin1 wild‐type mice. In vitro osteoclast forming assays with bone marrow‐derived monocyte/macrophage revealed that Pin1‐deficient osteoclasts are larger than wild‐type osteoclasts and have higher nuclei numbers, indicating greater extent of fusion. Pin1 deficiency also highly enhanced foreign body giant cell formation both in vitro and in vivo. Among the known fusion proteins, only DC‐STAMP was significantly increased in Pin1−/− osteoclasts. Immunohistochemistry showed that DC‐STAMP expression was also significantly increased in tibial metaphysis of Pin1 KO mice. We found that Pin1 binds and isomerizes DC‐STAMP and affects its expression levels and localization at the plasma membrane. Taken together, our data indicate that Pin1 is a determinant of bone mass through the regulation of the osteoclast fusion protein DC‐STAMP. The identification of Pin1 as a factor involved in cell fusion contributes to the understanding of osteoclast‐associated diseases, including osteoporosis, and opens new avenues for therapeutic targets. J. Cell. Physiol. 229: 2166–2174, 2014.


Journal of Cellular Physiology | 2013

Pin1-mediated Runx2 modification is critical for skeletal development

Won-Joon Yoon; Rabia Islam; Young Dan Cho; Kyung Mi Woo; Jeong Hwa Baek; Takafumi Uchida; Toshihisa Komori; Andre J. van Wijnen; Janet L. Stein; Jane B. Lian; Gary S. Stein; Je Yong Choi; Suk Chul Bae; Hyun-Mo Ryoo

Runx2 is the master transcription factor for bone formation. Haploinsufficiency of RUNX2 is the genetic cause of cleidocranial dysplasia (CCD) that is characterized by hypoplastic clavicles and open fontanels. In this study, we found that Pin1, peptidyl prolyl cis–trans isomerase, is a critical regulator of Runx2 in vivo and in vitro. Pin1 mutant mice developed CCD‐like phenotypes with hypoplastic clavicles and open fontanels as found in the Runx2+/− mice. In addition Runx2 protein level was significantly reduced in Pin1 mutant mice. Moreover Pin1 directly interacts with the Runx2 protein in a phosphorylation‐dependent manner and subsequently stabilizes Runx2 protein. In the absence of Pin1, Runx2 is rapidly degraded by the ubiquitin‐dependent protein degradation pathway. However, Pin1 overexpression strongly attenuated uniquitin‐dependent Runx2 degradation. Collectively conformational change of Runx2 by Pin1 is essential for its protein stability and possibly enhances the level of active Runx2 in vivo. J. Cell. Physiol. 228: 2377–2385, 2013.


Journal of Biological Chemistry | 2016

Pin1-mediated Modification Prolongs the Nuclear Retention of β-Catenin in Wnt3a-induced Osteoblast Differentiation.

Hye-Rim Shin; Rabia Islam; Won-Joon Yoon; Taegyung Lee; Young-Dan Cho; Han-Sol Bae; B. Kim; Kyung Mi Woo; Jeong-Hwa Baek; Hyun-Mo Ryoo

The canonical Wnt signaling pathway, in which β-catenin nuclear localization is a crucial step, plays an important role in osteoblast differentiation. Pin1, a prolyl isomerase, is also known as a key enzyme in osteogenesis. However, the role of Pin1 in canonical Wnt signal-induced osteoblast differentiation is poorly understood. We found that Pin1 deficiency caused osteopenia and reduction of β-catenin in bone lining cells. Similarly, Pin1 knockdown or treatment with Pin1 inhibitors strongly decreased the nuclear β-catenin level, TOP flash activity, and expression of bone marker genes induced by canonical Wnt activation and vice versa in Pin1 overexpression. Pin1 interacts directly with and isomerizes β-catenin in the nucleus. The isomerized β-catenin could not bind to nuclear adenomatous polyposis coli, which drives β-catenin out of the nucleus for proteasomal degradation, which consequently increases the retention of β-catenin in the nucleus and might explain the decrease of β-catenin ubiquitination. These results indicate that Pin1 could be a critical target to modulate β-catenin-mediated osteogenesis.


Journal of Cellular Physiology | 2015

Pin1 plays a critical role as a molecular switch in canonical BMP signaling.

Won-Joon Yoon; Rabia Islam; Young-Dan Cho; Kyung-Min Ryu; Hye-Rim Shin; Kyung Mi Woo; Jeong-Hwa Baek; Hyun-Mo Ryoo

Pin1 is a peptidyl prolyl cis‐trans isomerase that specifically binds to the phosphoserine–proline or phosphothreonine–proline motifs of numerous proteins. Previously, we reported that Pin1 deficiency resulted in defects in osteoblast differentiation during early bone development. In this study, we found that adult Pin1‐deficient mice developed osteoporotic phenotypes compared to age‐matched controls. Since BMP2 stored in the bone matrix plays a critical role in adult bone maintenance, we suspected that BMP R‐Smads (Smad1 and Smad5) could be critical targets for Pin1 action. Pin1 specifically binds to the phosphorylated linker region of Smad1, which leads to structural modification and stabilization of the Smad1 protein. In this process, Pin1‐mediated conformational modification of Smad1 directly suppresses the Smurf1 interaction with Smad1, thereby promoting sustained activation of the Smad1 molecule. Our data demonstrate that post‐phosphorylational prolyl isomerization of Smad1 is a converging signal to stabilize the Smad1 molecule against the ubiquitination process mediated by Smurf1. Therefore, Pin1 is a critical molecular switch in the determination of Smad1 fate, opposing the death signal transmitted to the Smad1 linker region by phosphorylation cascades after its nuclear localization and transcriptional activation. Thus, Pin1 could be developed as a major therapeutic target in many skeletal diseases. J. Cell. Physiol. 230: 640–647, 2015.


Journal of Cellular Physiology | 2014

Pin1-Mediated Prolyl Isomerization of Runx1 Affects PU.1 Expression in Pre-Monocytes

Rabia Islam; Won-Joon Yoon; Kyung Mi Woo; Jeong-Hwa Baek; Hyun-Mo Ryoo

Regulation of the hematopoietic transcription factor PU.1, a member of the ETS family, plays a critical role in the development of blood cells and in leukemia. The dosage of PU.1 has been shown to cause a shift in myelomonocytic progenitor fate. Pin1 is a unique substrate‐specific enzyme that can isomerize phospho‐Ser/Thr–Pro peptide bonds, accelerating the conformational change in its substrates between a cis and a trans form. Such activity has been demonstrated to be a tightly controlled mechanism regulating a wide variety of protein functions under both normal physiological and pathological conditions. We have previously reported that a conformational change in Runx2 induced by Pin1 is essential for its function in osteogenesis in vitro and in vivo. In this study, we show that the Pin1‐mediated conformational change in Runx1 enhances its acetylation and stabilization and, consequently, enhances its transacting activity. The increased acetylation of Runx1 represses PU.1 transcription in pre‐monocytes. Conversely, the lack of (or the inhibition of) Pin1 increases PU.1 transcription in vitro and in vivo in pre‐monocytes and in the spleen tissue. Pin1 KO mice have an increased CD11b+/F4/80+ cell population and F4/80 protein expression in spleen. From our data, we can conclude that the conformational change in Runx1 induced by Pin1 represses PU.1 transcription in pre‐monocytes and influences the commitment to the monocyte lineage. The dosage of PU.1 is a crucial factor in acute myeloid leukemia (AML), and Pin1 may thus be a useful target for controlling PU.1‐dependent hematopoiesis, as well as leukemogenesis. J. Cell. Physiol. 229: 443–452, 2014.


Journal of Cellular Physiology | 2017

Pin1, the Master Orchestrator of Bone Cell Differentiation.

Rabia Islam; Won-Joon Yoon; Hyun-Mo Ryoo

Pin1 is an enzyme that specifically recognizes the peptide bond between phosphorylated serine or threonine (pS/pT‐P) and proline. This recognition causes a conformational change of its substrate, which further regulates downstream signaling. Pin1−/− mice show developmental bone defects and reduced mineralization. Pin1 targets RUNX2 (Runt‐Related Transcription Factor 2), SMAD1/5, and β‐catenin in the FGF, BMP, and WNT pathways, respectively. Pin1 has multiple roles in the crosstalk between different anabolic bone signaling pathways. For example, it controls different aspects of osteoblastogenesis and increases the transcriptional activity of Runx2, both directly and indirectly. Pin1 also influences osteoclastogenesis at different stages by targeting PU.1 (Purine‐rich nucleic acid binding protein 1), C‐FOS, and DC‐STAMP. The phenotype of Pin1−/− mice has led to the recent identification of multiple roles of Pin1 in different molecular pathways in bone cells. These roles suggest that Pin1 can be utilized as an efficient drug target in congenital and acquired bone diseases. J. Cell. Physiol. 232: 2339–2347, 2017.


Journal of Bone and Mineral Research | 2017

An HDAC Inhibitor, Entinostat/MS-275, Partially Prevents Delayed Cranial Suture Closure in Heterozygous Runx2 Null Mice

Han-Sol Bae; Won-Joon Yoon; Young-Dan Cho; Rabia Islam; Hye-Rim Shin; Bong-Soo Kim; Jin-Muk Lim; Minseok Seo; Seoae Cho; Kang-Young Choi; Seung-Hak Baek; Hong-Gee Kim; Kyung Mi Woo; Jeong-Hwa Baek; Yun-Sil Lee; Hyun-Mo Ryoo

Cleidocranial dysplasia (CCD) is an autosomal dominant skeletal disorder caused by mutations in RUNX2, coding a key transcription factor of early osteogenesis. CCD patients suffer from developmental defects in cranial bones. Despite numerous investigations and clinical approaches, no therapeutic strategy has been suggested to prevent CCD. Here, we show that fetal administration of Entinostat/MS‐275, a class I histone deacetylase (HDAC)‐specific inhibitor, partially prevents delayed closure of cranial sutures in Runx2+/‐ mice strain of C57BL/6J by two mechanisms: 1) posttranslational acetylation of Runx2 protein, which stabilized the protein and activated its transcriptional activity; and 2) epigenetic regulation of Runx2 and other bone marker genes. Moreover, we show that MS‐275 stimulates osteoblast proliferation effectively both in vivo and in vitro, suggesting that delayed skeletal development in CCD is closely related to the decreased number of progenitor cells as well as the delayed osteogenic differentiation. These findings provide the potential benefits of the therapeutic strategy using MS‐275 to prevent CCD.


Journal of Cellular Physiology | 2018

Peptidyl-prolyl cis-trans isomerase NIMA interacting 1 regulates skeletal muscle fusion through structural modification of Smad3 in the linker region: ISLAM et al.

Rabia Islam; Heein Yoon; Hye-Rim Shin; Han-Sol Bae; Bong-Soo Kim; Won-Joon Yoon; Kyung Mi Woo; Jeong-Hwa Baek; Yun-Sil Lee; Hyun-Mo Ryoo

Myoblast fusion is critical for muscle growth, regeneration, and repair. We previously reported that the enzyme peptidyl‐prolyl cis–trans isomerase NIMA interacting 1 (Pin1) is involved in osteoclast fusion. The objective of this study was to investigate the possibility that Pin1 also inhibits myoblast fusion. Here, we show the increased number of nuclei in the Pin1+/− mice muscle fiber compared to that in wild‐type mice. Moreover, we show that low dose of the Pin1 inhibitor dipentamethylene thiuram monosulfide treatment caused enhanced fusion in C2C12 cells. The R‐Smads are well‐known mediators of muscle hypertrophy and hyperplasia as well as being substrates of Pin1. We found that Pin1 is crucial for maintaining the stability of Smad3 (homologues of the Drosophila protein, mothers against decapentaplegic (Mad) and the Caenorhabditis elegans protein Sma). Our results show that serine 204 within Smad3 is the key Pin1‐binding site during inhibition of myoblast fusion and that both the transforming growth factor‐β receptor and extracellular signal‐regulated kinase (ERK)‐mediated phosphorylation are required for the interaction of Pin1 with Smad3. These findings suggest that a precise level of Pin1 activity is essential for regulating myoblast fusion during myogenesis and muscle regeneration.


Scientific Reports | 2017

Blood-testis barrier integrity depends on Pin1 expression in Sertoli cells

Rabia Islam; Heein Yoon; Bong-Soo Kim; Han-Sol Bae; Hye-Rim Shin; Woo-Jin Kim; Won-Joon Yoon; Yun-Sil Lee; Kyung Mi Woo; Jeong-Hwa Baek; Hyun-Mo Ryoo

The conformation and function of a subset of serine and threonine-phosphorylated proteins are regulated by the prolyl isomerase Pin1 through isomerization of phosphorylated Ser/Thr-Pro bonds. Pin1 is intensely expressed in Sertoli cells, but its function in this post mitotic cell remains unclear. Our aim was to investigate the role of Pin1 in the Sertoli cells. Lack of Pin1 caused disruption of the blood-testis barrier. We next investigated if the activin pathways in the Sertoli cells were affected by lack of Pin1 through immunostaining for Smad3 protein in testis tissue. Indeed, lack of Pin1 caused reduced Smad3 expression in the testis tissue, as well as a reduction in the level of N-Cadherin, a known target of Smad3. Pin1−/− testes express Sertoli cell marker mRNAs in a pattern similar to that seen in Smad3+/− mice, except for an increase in Wt1 expression. The resulting dysregulation of N-Cadherin, connexin 43, and Wt1 targets caused by lack of Pin1 might affect the mesenchymal–epithelial balance in the Sertoli cells and perturb the blood-testis barrier. The effect of Pin1 dosage in Sertoli cells might be useful in the study of toxicant-mediated infertility, gonadal cancer, and for designing male contraceptives.

Collaboration


Dive into the Rabia Islam's collaboration.

Top Co-Authors

Avatar

Hyun-Mo Ryoo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Won-Joon Yoon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kyung Mi Woo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Hwa Baek

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Han-Sol Bae

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hye-Rim Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Young-Dan Cho

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Woo-Jin Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Yun-Sil Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge