Network


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

Hotspot


Dive into the research topics where Rafiquel Sarker is active.

Publication


Featured researches published by Rafiquel Sarker.


Nature Structural & Molecular Biology | 2002

Three-dimensional structure of a bacterial oxalate transporter.

Teruhisa Hirai; Jurgen Heymann; Dan Shi; Rafiquel Sarker; Peter C. Maloney; Sriram Subramaniam

The major facilitator superfamily (MFS) represents one of the largest classes of evolutionarily related membrane transporter proteins. Here we present the three-dimensional structure at 6.5 Å resolution of a bacterial member of this superfamily, OxlT. The structure, derived from an electron crystallographic analysis of two-dimensional crystals, reveals that the 12 helices in the OxlT molecule are arranged around a central cavity, which is widest at the center of the membrane. The helices divide naturally into three groups: a peripheral set comprising helices 3, 6, 9 and 12; a second set comprising helices 2, 5, 8 and 11 that faces the central substrate transport pathway across most of the length of the membrane; and a third set comprising helices 1, 4, 7 and 10 that participate in the pathway either on the cytoplasmic side (4 and 10) or on the periplasmic side (1 and 7). Overall, the architecture of the protein is remarkably symmetric, providing a compelling molecular explanation for the ability of such transporters to carry out bi-directional substrate transport.


The Journal of Experimental Biology | 2009

NHE3 regulatory complexes

Mark Donowitz; Sachin Mohan; Cindy Xinjun Zhu; Tian E. Chen; Rong Lin; Boyoung Cha; Nicholas C. Zachos; Rakhilya Murtazina; Rafiquel Sarker; Xuhang Li

SUMMARY The epithelial brush border Na/H exchanger NHE3 is active under basal conditions and functions as part of neutral NaCl absorption in the intestine and renal proximal tubule, where it accounts for the majority of total Na absorbed. NHE3 is highly regulated. Both stimulation and inhibition occur post-prandially. This digestion related regulation of NHE3 is mimicked by multiple extracellular agonists and intracellular second messengers. The regulation of NHE3 depends on its C-terminal cytoplasmic domain, which acts as a scaffold to bind multiple regulatory proteins and links NHE3 to the cytoskeleton. The cytoskeletal association occurs by both direct binding to ezrin and by indirect binding via ezrin binding to the C-terminus of the multi-PDZ domain containing proteins NHERF1 and NHERF2. This is a review of the domain structure of NHE3 and of the scaffolding function and role in the regulation of NHE3 of the NHE3 C-terminal domain.


Gastroenterology | 2011

D-Glucose Acts via Sodium/Glucose Cotransporter 1 to Increase NHE3 in Mouse Jejunal Brush Border by a Na+/H+ Exchange Regulatory Factor 2–Dependent Process

Rong Lin; Rakhilya Murtazina; Boyoung Cha; Molee Chakraborty; Rafiquel Sarker; Tian E. Chen; Zhihong Lin; Boris M. Hogema; Hugo R. de Jonge; Ursula Seidler; Jerrold R. Turner; Xuhang Li; Olga Kovbasnjuk; Mark Donowitz

BACKGROUND & AIMS Oral rehydration solutions reduce diarrhea-associated mortality. Stimulated sodium absorption by these solutions is mediated by the Na(+)/H(+) hydrogen exchanger NHE3 and is increased by Na(+)-glucose co-transport in vitro, but the mechanisms of this up-regulated process are only partially understood. METHODS Intracellular pH was measured in jejunal enterocytes of wild-type mice and mice with disrupted Na+/H+ exchange regulatory co-factor 2 (NHERF2-/- mice) by multiphoton microscopy. Diarrhea was induced by cholera toxin. Caco-2BBe cells that express NHE3 and the sodium/glucose cotransporter 1 (SGLT1) were studied by fluorometry, before and after siRNA-mediated knockdown of NHERF1 or NHERF2. NHE3 distribution was assessed by cell-surface biotinylation and confocal microscopy. Brush-border mobility was determined by fluorescence recovery after photobleaching and confocal microscopy. RESULTS The nonmetabolized SGLT1 substrate α-methyl-D-Glu (α-MD-G) activated jejunal NHE3; this process required Akt and NHERF2. α-MD-G normalized NHE3 activity after cholera toxin-induced diarrhea. α-MD-G-stimulated jejunal NHE3 activity was defective in NHERF2-/- mice and cells with NHERF2 knockdown, but occurred normally with NHERF1 knockdown; was associated with increased NHE3 surface expression in Caco-2 cells, which also was NHERF2-dependent; was associated with dissociation of NHE3 from NHERF2 and an increase in the NHE3 mobile fraction from the brush border; and was accompanied by a NHERF2 ezrin-radixin-moesin-binding domain-dependent increase in co-precipitation of ezrin with NHE3. CONCLUSIONS SGLT1-mediated Na-glucose co-transport stimulates NHE3 activity in vivo by an Akt- and NHERF2-dependent signaling pathway. It is associated with increased brush-border NHE3 and association between ezrin and NHE3. Activation of NHE3 corrects cholera toxin-induced defects in Na absorption and might contribute to the efficacy of oral rehydration solutions.


The EMBO Journal | 2001

Projection structure and molecular architecture of OxlT, a bacterial membrane transporter.

Jurgen Heymann; Rafiquel Sarker; Teruhisa Hirai; Dan Shi; Jacqueline L. S. Milne; Peter C. Maloney; Sriram Subramaniam

The major facilitator superfamily (MFS) represents the largest collection of evolutionarily related members within the class of membrane ‘carrier’ proteins. OxlT, a representative example of the MFS, is an oxalate‐transporting membrane protein in Oxalobacter formigenes. From an electron crystallographic analysis of two‐dimensional crystals of OxlT, we have determined the projection structure of this membrane transporter. The projection map at 6 Å resolution indicates the presence of 12 transmembrane helices in each monomer of OxlT, with one set of six helices related to the other set by an approximate internal two‐fold axis. The projection map reveals the existence of a central cavity, which we propose to be part of the pathway of oxalate transport. By combining information from the projection map with related biochemical data, we present probable models for the architectural arrangement of transmembrane helices in this protein superfamily.


Molecular Biology of the Cell | 2008

Casein Kinase 2 Binds to the C Terminus of Na+/H+ exchanger 3 (NHE3) and Stimulates NHE3 Basal Activity by Phosphorylating a Separate Site in NHE3

Rafiquel Sarker; Mads Grønborg; Boyoung Cha; Sachin Mohan; Yueping Chen; Akhilesh Pandey; David W. Litchfield; Mark Donowitz; Xuhang Li

Na(+)/H(+) exchanger 3 (NHE3) is the epithelial-brush border isoform responsible for most intestinal and renal Na(+) absorption. Its activity is both up- and down-regulated under normal physiological conditions, and it is inhibited in most diarrheal diseases. NHE3 is phosphorylated under basal conditions and Ser/Thr phosphatase inhibitors stimulate basal exchange activity; however, the kinases involved are unknown. To identify kinases that regulate NHE3 under basal conditions, NHE3 was immunoprecipitated; LC-MS/MS of trypsinized NHE3 identified a novel phosphorylation site at S(719) of the C terminus, which was predicted to be a casein kinase 2 (CK2) phosphorylation site. This was confirmed by an in vitro kinase assay. The NHE3-S719A mutant but not NHE3-S719D had reduced NHE3 activity due to less plasma membrane NHE3. This was due to reduced exocytosis plus decreased plasma membrane delivery of newly synthesized NHE3. Also, NHE3 activity was inhibited by the CK2 inhibitor 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole DMAT when wild-type NHE3 was expressed in fibroblasts and Caco-2 cells, but the NHE3-S(719) mutant was fully resistant to DMAT. CK2 bound to the NHE3 C-terminal domain, between amino acids 590 and 667, a site different from the site it phosphorylates. CK2 binds to the NHE3 C terminus and stimulates basal NHE3 activity by phosphorylating a separate single site on the NHE3 C terminus (S(719)), which affects NHE3 trafficking.


Human Molecular Genetics | 2015

Reduced sodium/proton exchanger NHE3 activity causes congenital sodium diarrhea

Andreas R. Janecke; Peter Heinz-Erian; Jianyi Yin; Britt-Sabina Petersen; Andre Franke; Silvia Lechner; Irene Fuchs; Serge Melancon; Holm H. Uhlig; Simon Travis; Evelyne Marinier; Vojislav N. Perisic; Nina Ristic; Patrick Gerner; I W Booth; Satu Wedenoja; Nadja Baumgartner; Julia Vodopiutz; Marie Christine Frechette-Duval; Jan De Lafollie; Rabindranath Persad; Neil Warner; C. Ming Tse; Karan Sud; Nicholas C. Zachos; Rafiquel Sarker; Xinjun Zhu; Aleixo M. Muise; Klaus Peter Zimmer; Heiko Witt

Congenital sodium diarrhea (CSD) refers to an intractable diarrhea of intrauterine onset with high fecal sodium loss. CSD is clinically and genetically heterogeneous. Syndromic CSD is caused by SPINT2 mutations. While we recently described four cases of the non-syndromic form of CSD that were caused by dominant activating mutations in intestinal receptor guanylate cyclase C (GC-C), the genetic cause for the majority of CSD is still unknown. Therefore, we aimed to determine the genetic cause for non-GC-C non-syndromic CSD in 18 patients from 16 unrelated families applying whole-exome sequencing and/or chromosomal microarray analyses and/or direct Sanger sequencing. SLC9A3 missense, splicing and truncation mutations, including an instance of uniparental disomy, and whole-gene deletion were identified in nine patients from eight families with CSD. Two of these nine patients developed inflammatory bowel disease (IBD) at 4 and 16 years of age. SLC9A3 encodes Na(+)/H(+) antiporter 3 (NHE3), which is the major intestinal brush-border Na(+)/H(+) exchanger. All mutations were in the NHE3 N-terminal transport domain, and all missense mutations were in the putative membrane-spanning domains. Identified SLC9A3 missense mutations were functionally characterized in plasma membrane NHE null fibroblasts. SLC9A3 missense mutations compromised NHE3 activity by reducing basal surface expression and/or loss of basal transport function of NHE3 molecules, whereas acute regulation was normal. This study identifies recessive mutations in NHE3, a downstream target of GC-C, as a cause of CSD and implies primary basal NHE3 malfunction as a predisposition for IBD in a subset of patients.


American Journal of Physiology-cell Physiology | 2011

NHERF1 and NHERF2 are necessary for multiple but usually separate aspects of basal and acute regulation of NHE3 activity

Rafiquel Sarker; Vera E. Valkhoff; Nicholas C. Zachos; Rong Lin; Boyoung Cha; Tian E. Chen; Sandra E. Guggino; Mirza Zizak; Hugo R. de Jonge; Boris M. Hogema; Mark Donowitz

Na(+)/H(+) exchanger 3 (NHE3) is expressed in the brush border (BB) of intestinal epithelial cells and accounts for the majority of neutral NaCl absorption. It has been shown that the Na(+)/H(+) exchanger regulatory factor (NHERF) family members of multi-PDZ domain-containing scaffold proteins bind to the NHE3 COOH terminus and play necessary roles in NHE3 regulation in intestinal epithelial cells. Most studies of NHE3 regulation have been in cell models in which NHERF1 and/or NHERF2 were overexpressed. We have now developed an intestinal Na(+) absorptive cell model in Caco-2/bbe cells by expressing hemagglutinin (HA)-tagged NHE3 with an adenoviral infection system. Roles of NHERF1 and NHERF2 in NHE3 regulation were determined, including inhibition by cAMP, cGMP, and Ca(2+) and stimulation by EGF, with knockdown (KD) approaches with lentivirus (Lenti)-short hairpin RNA (shRNA) and/or adenovirus (Adeno)-small interfering RNA (siRNA). Stable infection of Caco-2/bbe cells by NHERF1 or NHERF2 Lenti-shRNA significantly and specifically reduced NHERF protein expression by >80%. NHERF1 KD reduced basal NHE3 activity, while NHERF2 KD stimulated NHE3 activity. siRNA-mediated (transient) and Lenti-shRNA-mediated (stable) gene silencing of NHERF2 (but not of NHERF1) abolished cGMP- and Ca(2+)-dependent inhibition of NHE3. KD of NHERF1 or NHERF2 alone had no effect on cAMP inhibition of NHE3, but KD of both simultaneously abolished the effect of cAMP. The stimulatory effect of EGF on NHE3 was eliminated in NHERF1-KD but occurred normally in NHERF2-KD cells. These findings show that both NHERF2 and NHERF1 are involved in setting NHE3 activity. NHERF2 is necessary for cGMP-dependent protein kinase (cGK) II- and Ca(2+)-dependent inhibition of NHE3. cAMP-dependent inhibition of NHE3 activity requires either NHERF1 or NHERF2. Stimulation of NHE3 activity by EGF is NHERF1 dependent.


Gut | 2014

Farnesoid X receptor agonists attenuate colonic epithelial secretory function and prevent experimental diarrhoea in vivo

Magdalena S. Mroz; Niamh Keating; Joseph B. Ward; Rafiquel Sarker; Silvie Amu; Gabriella Aviello; Mark Donowitz; Padraic G. Fallon; Stephen J. Keely

Objective Bile acids are important regulators of intestinal physiology, and the nuclear bile acid receptor, farnesoid X receptor (FXR), is emerging as a promising therapeutic target for several intestinal disorders. Here, we investigated a role for FXR in regulating intestinal fluid and electrolyte transport and the potential for FXR agonists in treating diarrhoeal diseases. Design Electrogenic ion transport was measured as changes in short-circuit current across voltage-clamped T84 cell monolayers or mouse tissues in Ussing chambers. NHE3 activity was measured as BCECF fluorescence in Caco-2 cells. Protein expression was measured by immunoblotting and cell surface biotinylation. Antidiarrhoeal efficacy of GW4064 was assessed using two in vivo mouse models: the ovalbumin-induced diarrhoea model and cholera toxin (CTX)-induced intestinal fluid accumulation. Results GW4064 (5 μmol/L; 24 h), a specific FXR agonist, induced nuclear translocation of the receptor in T84 cells and attenuated Cl− secretory responses to both Ca2+ and cAMP-dependent agonists. GW4064 also prevented agonist-induced inhibition of NHE3 in Caco-2 cells. In mice, intraperitoneal administration of GW4064 (50 mg/mL) also inhibited Ca2+ and cAMP-dependent secretory responses across ex vivo colonic tissues and prevented ovalbumin-induced diarrhoea and CTX-induced intestinal fluid accumulation in vivo. At the molecular level, FXR activation attenuated apical Cl− currents by inhibiting expression of cystic fibrosis transmembrane conductance regulator channels and inhibited basolateral Na+/K+-ATPase activity without altering expression of the protein. Conclusions These data reveal a novel antisecretory role for the FXR in colonic epithelial cells and suggest that FXR agonists have excellent potential for development as a new class of antidiarrheal drugs.


Journal of Biological Chemistry | 2009

NHERF3 (PDZK1) Contributes to Basal and Calcium Inhibition of NHE3 Activity in Caco-2BBe Cells

Nicholas C. Zachos; Xuhang Li; Olga Kovbasnjuk; Boris M. Hogema; Rafiquel Sarker; Luke J. Lee; Min Li; Hugo R. de Jonge; Mark Donowitz

Elevated intracellular Ca2+ ([Ca2+]i) inhibition of NHE3 is reconstituted by NHERF2, but not NHERF1, by a mechanism involving the formation of multiprotein signaling complexes. To further evaluate the specificity of the NHERF family in calcium regulation of NHE3 activity, the current study determined whether NHERF3 reconstitutes elevated [Ca2+]i regulation of NHE3. In vitro, NHERF3 bound the NHE3 C terminus between amino acids 588 and 667. In vivo, NHE3 and NHERF3 associate under basal conditions as indicated by co-immunoprecipitation, confocal microscopy, and fluorescence resonance energy transfer. Treatment of PS120/NHE3/NHERF3 cells, but not PS120/NHE3 cells, with the Ca2+ ionophore, 4-bromo-A23187 (0.5 μm): 1) inhibited NHE3 Vmax activity; 2) decreased NHE3 surface amount; 3) dissociated NHE3 and NHERF3 at the plasma membrane by confocal immunofluorescence and fluorescence resonance energy transfer. Similarly, in Caco-2BBe cells, NHERF3 and NHE3 colocalized in the BB under basal conditions but after elevation of [Ca2+]i by carbachol, this overlap was abolished. NHERF3 short hairpin RNA knockdown (>50%) in Caco-2BBe cells significantly reduced basal NHE3 activity by decreasing BB NHE3 amount. Also, carbachol-mediated inhibition of NHE3 activity was abolished in Caco-2BBe cells in which NHERF3 protein expression was significantly reduced. In summary: 1) NHERF3 colocalizes and directly binds NHE3 at the plasma membrane under basal conditions; 2) NHERF3 reconstitutes [Ca2+]i inhibition of NHE3 activity and dissociates from NHE3 in fibroblasts and polarized intestinal epithelial cells with elevated [Ca2+]i; 3) NHERF3 short hairpin RNA significantly reduced NHE3 basal activity and brush border expression in Caco-2BBe cells. These results demonstrate that NHERF3 reconstitutes calcium inhibition of NHE3 activity by anchoring NHE3 basally and releasing it with elevated Ca2+.


Annals of the New York Academy of Sciences | 2009

A New Insight into Pathophysiological Mechanisms of Zinc in Diarrhea

Kazi Mirajul Hoque; Rafiquel Sarker; Sandra E. Guggino; Chung Ming Tse

An increasing amount of data showing the beneficial use of zinc (Zn) in treating diarrhea continues to emerge from epidemiological and clinical trials. However, without a thorough understanding of physiological mechanisms of Zn, it does not support policy recommendation to advocate the therapeutic use of Zn. Our data demonstrate that Zn is a potential antidiarrheal agent that provides substantial benefit by stimulating sodium absorption and inhibiting chloride secretion in intestinal epithelial cells. Thus, inclusion of Zn in oral rehydration solution (ORS) has the potential to markedly augment the effectiveness of ORS in the treatment of diarrhea.

Collaboration


Dive into the Rafiquel Sarker's collaboration.

Top Co-Authors

Avatar

Mark Donowitz

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Boyoung Cha

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Nicholas C. Zachos

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jianbo Yang

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Olga Kovbasnjuk

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tian-e Chen

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ming Tse

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rakhilya Murtazina

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tian E. Chen

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xuhang Li

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge