Network


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

Hotspot


Dive into the research topics where Chinmoy Patra is active.

Publication


Featured researches published by Chinmoy Patra.


Biomaterials | 2012

Silk protein fibroin from Antheraea mylitta for cardiac tissue engineering

Chinmoy Patra; Sarmistha Talukdar; Tatyana Novoyatleva; Siva R. Velagala; Christian Mühlfeld; Banani Kundu; Subhas C. Kundu; Felix B. Engel

The human heart cannot regenerate after an injury. Lost cardiomyocytes are replaced by scar tissue resulting in reduced cardiac function causing high morbidity and mortality. One possible solution to this problem is cardiac tissue engineering. Here, we have investigated the suitability of non-mulberry silk protein fibroin from Indian tropical tasar Antheraea mylitta as a scaffold for engineering a cardiac patch in vitro. We have tested cell adhesion, cellular metabolic activity, response to extracellular stimuli, cell-to-cell communication and contractility of 3-days postnatal rat cardiomyocytes on silk fibroin. Our data demonstrate that A. mylitta silk fibroin exhibits similar properties as fibronectin, a component of the natural matrix for cardiomyocytes. Comparison to mulberry Bombyx mori silk protein fibroin shows that A. mylitta silk fibroin is superior probably due to its RGD domains. 3D scaffolds can efficiently be loaded with cardiomyocytes resulting in contractile patches. In conclusion, our findings demonstrate that A. mylitta silk fibroin 3D scaffolds are suitable for the engineering of cardiac patches.


The Journal of Neuroscience | 2013

Gpr126 functions in Schwann cells to control differentiation and myelination via G-protein activation.

Amit Mogha; Andrew E. Benesh; Chinmoy Patra; Felix B. Engel; Torsten Schöneberg; Ines Liebscher; Kelly R. Monk

The myelin sheath surrounding axons ensures that nerve impulses travel quickly and efficiently, allowing for the proper function of the vertebrate nervous system. We previously showed that the adhesion G-protein-coupled receptor (aGPCR) Gpr126 is essential for peripheral nervous system myelination, although the molecular mechanisms by which Gpr126 functions were incompletely understood. aGPCRs are a significantly understudied protein class, and it was unknown whether Gpr126 couples to G-proteins. Here, we analyze DhhCre;Gpr126fl/fl conditional mutants, and show that Gpr126 functions in Schwann cells (SCs) for radial sorting of axons and myelination. Furthermore, we demonstrate that elevation of cAMP levels or protein kinase A activation suppresses myelin defects in Gpr126 mouse mutants and that cAMP levels are reduced in conditional Gpr126 mutant peripheral nerve. Finally, we show that GPR126 directly increases cAMP by coupling to heterotrimeric G-proteins. Together, these data support a model in which Gpr126 functions in SCs for proper development and myelination and provide evidence that these functions are mediated via G-protein-signaling pathways.


Cardiovascular Research | 2010

TWEAK is a positive regulator of cardiomyocyte proliferation

Tatyana Novoyatleva; Florian Diehl; Machteld J. van Amerongen; Chinmoy Patra; Fulvia Ferrazzi; Riccardo Bellazzi; Felix B. Engel

AIMS Proliferation of mammalian cardiomyocytes stops during the first weeks after birth, preventing the heart from regenerating after injury. Recently, several studies have indicated that induction of cardiomyocyte proliferation can be utilized to regenerate the mammalian heart. Thus, it is important to identify novel factors that can induce proliferation of cardiomyocytes. Here, we determine the effect of TNF-related weak inducer of apoptosis (TWEAK) on cardiomyocytes, a cytokine known to regulate proliferation in several other cell types. METHODS AND RESULTS Stimulation of neonatal rat cardiomyocytes with TWEAK resulted in increased DNA synthesis, increased expression of the proliferative markers Cyclin D2 and Ki67, and downregulation of the cell cycle inhibitor p27KIP1. Importantly, TWEAK stimulation resulted also in mitosis (H3P), cytokinesis (Aurora B), and increased cardiomyocyte numbers. Loss of function experiments revealed that re-induction of proliferation was dependent on tumour necrosis factor receptor superfamily member 12A (FN14) signalling. Downstream signalling was mediated through activation of extracellular signal-regulated kinases and phosphatidylinositol 3-kinase as well as inhibition of glycogen synthase kinase-3beta. In contrast to neonatal cardiomyocytes, TWEAK had no effect on adult rat cardiomyocytes due to developmental downregulation of its receptor FN14. However, adenoviral expression of FN14 enabled efficient induction of cell cycle re-entry in adult cardiomyocytes after TWEAK stimulation. CONCLUSION Our data establish TWEAK as a positive regulator of cardiomyocyte proliferation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Organ-specific function of adhesion G protein-coupled receptor GPR126 is domain-dependent

Chinmoy Patra; Machteld J. van Amerongen; Subhajit Ghosh; Filomena Ricciardi; Amna Sajjad; Tatyana Novoyatleva; Amit Mogha; Kelly R. Monk; Christian Mühlfeld; Felix B. Engel

Significance Adhesion G protein-coupled receptors (GPCRs) are expressed in many developing organs, immune cells, and cancer cells, suggesting that they might play an important role in physiological and pathological functions. Compared with their potential importance, their function and signaling mechanisms are poorly understood. Disruption of the G protein-coupled receptor 126 (Gpr126) gene in mice leads to lack of myelination in the peripheral nervous system (PNS) and heart abnormalities. Similarly, the zebrafish mutant line gpr126st49 exhibits PNS abnormalities but, in contrast, no heart phenotype. Here we provide an explanation for these discrepancies. The presented data suggest that in the heart, the N-terminal fragment of Gpr126 can act independently as a ligand or coreceptor. Taken together, our data provide evidence of tissue- and domain-specific adhesion GPCR function. Despite their abundance and multiple functions in a variety of organ systems, the function and signaling mechanisms of adhesion G protein-coupled receptors (GPCRs) are poorly understood. Adhesion GPCRs possess large N termini containing various functional domains. In addition, many of them are autoproteolytically cleaved at their GPS sites into an N-terminal fragment (NTF) and C-terminal fragment. Here we demonstrate that Gpr126 is expressed in the endocardium during early mouse heart development. Gpr126 knockout in mice and knockdown in zebrafish caused hypotrabeculation and affected mitochondrial function. Ectopic expression of Gpr126-NTF that lacks the GPS motif (NTFΔGPS) in zebrafish rescued the trabeculation but not the previously described myelination phenotype in the peripheral nervous system. These data support a model in which the NTF of Gpr126, in contrast to the C-terminal fragment, plays an important role in heart development. Collectively, our analysis provides a unique example of the versatile function and signaling properties of adhesion GPCRs in vertebrates.


Blood | 2013

EGFL7 ligates αvβ3 integrin to enhance vessel formation

Iva Nikolic; Nevenka Dudvarski Stanković; Frank Bicker; Jeannette Meister; Helene Braun; Khader Awwad; Jan Baumgart; Kirsten Simon; Serge C. Thal; Chinmoy Patra; Patrick N. Harter; Karl H. Plate; Felix B. Engel; Stefanie Dimmeler; Johannes A. Eble; Michel Mittelbronn; Michael K. E. Schäfer; Emmanouil Chavakis; Ingrid Fleming; Mirko H. H. Schmidt

Angiogenesis, defined as blood vessel formation from a preexisting vasculature, is governed by multiple signal cascades including integrin receptors, in particular integrin αVβ3. Here we identify the endothelial cell (EC)-secreted factor epidermal growth factor-like protein 7 (EGFL7) as a novel specific ligand of integrin αVβ3, thus providing mechanistic insight into its proangiogenic actions in vitro and in vivo. Specifically, EGFL7 attaches to the extracellular matrix and by its interaction with integrin αVβ3 increases the motility of EC, which allows EC to move on a sticky underground during vessel remodeling. We provide evidence that the deregulation of EGFL7 in zebrafish embryos leads to a severe integrin-dependent malformation of the caudal venous plexus, pointing toward the significance of EGFL7 in vessel development. In biopsy specimens of patients with neurologic diseases, vascular EGFL7 expression rose with increasing EC proliferation. Further, EGFL7 became upregulated in vessels of the stroke penumbra using a mouse model of reversible middle cerebral artery occlusion. Our data suggest that EGFL7 expression depends on the remodeling state of the existing vasculature rather than on the phenotype of neurologic disease analyzed. In sum, our work sheds a novel light on the molecular mechanism EGFL7 engages to govern physiological and pathological angiogenesis.


Science | 2016

Injury-induced ctgfa directs glial bridging and spinal cord regeneration in zebrafish

Mayssa H. Mokalled; Chinmoy Patra; Amy L. Dickson; Toyokazu Endo; Didier Y. R. Stainier; Kenneth D. Poss

Spinal cord regeneration in zebrafish Unlike humans, zebrafish can regenerate their spinal cord. Mokalled et al. identified a growth factor in zebrafish that helps this process (see the Perspective by Williams and He). The protein encoded by ctgfa (connective tissue growth factor a) is secreted after injury and encourages glial cells to form a bridge across the spinal lesion. Addition of this protein improved spinal cord repair in injured zebrafish. Science, this issue p. 630; see also p. 544 A connective tissue growth factor underlies the unusual regenerative capacity of the zebrafish central nervous system. Unlike mammals, zebrafish efficiently regenerate functional nervous system tissue after major spinal cord injury. Whereas glial scarring presents a roadblock for mammalian spinal cord repair, glial cells in zebrafish form a bridge across severed spinal cord tissue and facilitate regeneration. We performed a genome-wide profiling screen for secreted factors that are up-regulated during zebrafish spinal cord regeneration. We found that connective tissue growth factor a (ctgfa) is induced in and around glial cells that participate in initial bridging events. Mutations in ctgfa disrupted spinal cord repair, and transgenic ctgfa overexpression or local delivery of human CTGF recombinant protein accelerated bridging and functional regeneration. Our study reveals that CTGF is necessary and sufficient to stimulate glial bridging and natural spinal cord regeneration.


Development | 2011

Nephronectin regulates atrioventricular canal differentiation via Bmp4-Has2 signaling in zebrafish

Chinmoy Patra; Florian Diehl; Fulvia Ferrazzi; Machteld J. van Amerongen; Tatyana Novoyatleva; Liliana Schaefer; Christian Mühlfeld; Felix B. Engel

The extracellular matrix is crucial for organogenesis. It is a complex and dynamic component that regulates cell behavior by modulating the activity, bioavailability and presentation of growth factors to cell surface receptors. Here, we determined the role of the extracellular matrix protein Nephronectin (Npnt) in heart development using the zebrafish model system. The vertebrate heart is formed as a linear tube in which myocardium and endocardium are separated by a layer of extracellular matrix termed the cardiac jelly. During heart development, the cardiac jelly swells at the atrioventricular (AV) canal, which precedes valve formation. Here, we show that Npnt expression correlates with this process. Morpholino-mediated knockdown of Npnt prevents proper valve leaflet formation and trabeculation and results in greater than 85% lethality at 7 days post-fertilization. The earliest observed phenotype is an extended tube-like structure at the AV boundary. In addition, the expression of myocardial genes involved in cardiac valve formation (cspg2, fibulin 1, tbx2b, bmp4) is expanded and endocardial cells along the extended tube-like structure exhibit characteristics of AV cells (has2, notch1b and Alcam expression, cuboidal cell shape). Inhibition of has2 in npnt morphants rescues the endocardial, but not the myocardial, expansion. By contrast, reduction of BMP signaling in npnt morphants reduces the ectopic expression of myocardial and endocardial AV markers. Taken together, our results identify Npnt as a novel upstream regulator of Bmp4-Has2 signaling that plays a crucial role in AV canal differentiation.


Biomaterials | 2012

The functional properties of nephronectin: An adhesion molecule for cardiac tissue engineering

Chinmoy Patra; Filomena Ricciardi; Felix B. Engel

Despite significant advances in preventive cardiovascular medicine and therapy for acute and chronic heart failure, cardiovascular diseases remain among the leading causes of death worldwide. In recent years cardiac tissue engineering has been established as a possible future treatment option for cardiac disease. However, the quality of engineered myocardial tissues remains poor. In tissue engineering it is important that the scaffold allows cells to attach, spread, maintain their differentiation status or differentiate into functional cells in order to exhibit their physiological function. Here, we have investigated the suitability of the natural cardiac extracellular matrix component nephronectin as an adhesive material for cardiac tissue engineering. Primary neonatal rat cardiomyocytes were seeded on collagen-, fibronectin- or nephronectin-coated glass coverslips and analyzed for cell adhesion, cellular metabolic activity, response to extracellular stimuli, cell-to-cell communication, differentiation and contractility. Our data demonstrate that most neonatal cardiomyocytes attached in an RGD domain-dependent manner within 18 h to nephronectin. The cells exhibited high metabolic activity, responded to growth factor stimuli and maintained their differentiation status. Moreover, nephronectin promoted sarcomere maturation and alignment, cell-to-cell communication and synchronous contractions. In conclusion, our findings demonstrate that nephronectin has excellent properties for cardiomyocyte adhesion and function and thus has the potential to improve current cardiac tissue engineering approaches.


Thrombosis and Haemostasis | 2014

Vascularisation for cardiac tissue engineering: the extracellular matrix

Chinmoy Patra; Aldo R. Boccaccini; Felix B. Engel

Cardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 µm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial-based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.


The FASEB Journal | 2014

FGF1-mediated cardiomyocyte cell cycle reentry depends on the interaction of FGFR-1 and Fn14

Tatyana Novoyatleva; Amna Sajjad; Denys Pogoryelov; Chinmoy Patra; Ralph T. Schermuly; Felix B. Engel

Fibroblast growth factors (FGFs) signal through FGF receptors (FGFRs) mediating a broad range of cellular functions during embryonic development, as well as disease and regeneration during adulthood. Thus, it is important to understand the underlying molecular mechanisms that modulate this system. Here, we show that FGFR‐1 can interact with the TNF receptor superfamily member fibroblast growth factor‐inducible molecule 14 (Fn14) resulting in cardiomyocyte cell cycle reentry. FGF1‐induced cell cycle reentry in neonatal cardiomyocytes could be blocked by Fn14 inhibition, while TWEAK‐induced cell cycle activation was inhibited by blocking FGFR‐1 signaling. In addition, costimulation experiments revealed a synergistic effect of FGF1 and TWEAK in regard to cardiomyocyte cell cycle induction via PI3K/Akt signaling. Overexpression of Fn14 with either FGFR‐1 long [FGFR‐1(L)] or FGFR‐1 short [FGFR‐1(S)] isoforms resulted after FGF1/TWEAK stimulation in cell cycle reentry of >40% adult cardiomyocytes. Finally, coimmunoprecipitation and proximity ligation assays indicated that endogenous FGFR‐1 and Fn14 interact with each other in cardiomyocytes. This interaction was strongly enhanced in the presence of their corresponding ligands, FGF1 and TWEAK. Taken together, our data suggest that FGFR‐1/Fn14 interaction may represent a novel endogenous mechanism to modulate the action of these receptors and their ligands and to control cardiomyocyte cell cycle reentry.—Novoyatleva, T., Sajjad, A., Pogoryelov, D., Patra, C., Schermuly, R. T., Engel, F. B. FGF1‐mediated cardiomyocyte cell cycle reentry depends on the interaction of FGFR‐1 and Fn14. FASEB J. 28, 2492–2503 (2014). www.fasebj.org

Collaboration


Dive into the Chinmoy Patra's collaboration.

Top Co-Authors

Avatar

Felix B. Engel

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Florian Diehl

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kelly R. Monk

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amit Mogha

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge