Network


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

Hotspot


Dive into the research topics where Kalpana Nattamai is active.

Publication


Featured researches published by Kalpana Nattamai.


Nature | 2013

A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing

Maria Carolina Florian; Kalpana Nattamai; Karin Dörr; Gina Marka; Bettina Überle; Virag Vas; Christina Eckl; Immanuel Andrä; Matthias Schiemann; Robert A.J. Oostendorp; Karin Scharffetter-Kochanek; Hans A. Kestler; Yi Zheng; Hartmut Geiger

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid–lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Journal of Clinical Investigation | 2007

TNF-α induces leukemic clonal evolution ex vivo in Fanconi anemia group C murine stem cells

June Li; Daniel P. Sejas; Xiaoling Zhang; Yuhui Qiu; Kalpana Nattamai; Reena Rani; Keaney Rathbun; Hartmut Geiger; David A. Williams; Grover C. Bagby; Qishen Pang

The molecular pathogenesis of the myeloid leukemias that frequently occur in patients with Fanconi anemia (FA) is not well defined. Hematopoietic stem cells bearing inactivating mutations of FA complementation group C (FANCC) are genetically unstable and hypersensitive to apoptotic cytokine cues including IFN-γ and TNF-α, but neoplastic stem cell clones that arise frequently in vivo are resistant to these cytokines. Reasoning that the combination of genetic instability and cytokine hypersensitivity might create an environment supporting the emergence of leukemic stem cells, we tested the leukemia-promoting effects of TNF-α in murine stem cells. TNF-α exposure initially profoundly inhibited the growth of Fancc–/– stem cells. However, longer-term exposure of these cells promoted the outgrowth of cytogenetically abnormal clones that, upon transplantation into congenic WT mice, led to acute myelogenous leukemia. TNF-α induced ROS-dependent genetic instability in Fancc–/– but not in WT cells. The leukemic clones were TNF-α resistant but retained their characteristic hypersensitivity to mitomycin C and exhibited high levels of chromosomal instability. Expression of FANCC cDNA in Fancc–/– stem cells protected them from TNF-α–induced clonal evolution. We conclude that TNF-α exposure creates an environment in which somatically mutated preleukemic stem cell clones are selected and from which unaltered TNF-α–hypersensitive Fancc–/– stem cells are purged.


Nature Medicine | 2012

Pharmacological targeting of the thrombomodulin–activated protein C pathway mitigates radiation toxicity

Hartmut Geiger; Snehalata A Pawar; Edward J. Kerschen; Kalpana Nattamai; Irene Hernandez; Hai Po H. Liang; José A. Fernández; Jose A. Cancelas; Marnie A. Ryan; Olga Kustikova; Axel Schambach; Qiang Fu; Junru Wang; Louis M. Fink; Karl-Uwe Petersen; Daohong Zhou; Christopher Baum; Hartmut Weiler; Martin Hauer-Jensen

Tissue damage induced by ionizing radiation in the hematopoietic and gastrointestinal systems is the major cause of lethality in radiological emergency scenarios and underlies some deleterious side effects in patients undergoing radiation therapy. The identification of target-specific interventions that confer radiomitigating activity is an unmet challenge. Here we identify the thrombomodulin (Thbd)–activated protein C (aPC) pathway as a new mechanism for the mitigation of total body irradiation (TBI)-induced mortality. Although the effects of the endogenous Thbd-aPC pathway were largely confined to the local microenvironment of Thbd-expressing cells, systemic administration of soluble Thbd or aPC could reproduce and augment the radioprotective effect of the endogenous Thbd-aPC pathway. Therapeutic administration of recombinant, soluble Thbd or aPC to lethally irradiated wild-type mice resulted in an accelerated recovery of hematopoietic progenitor activity in bone marrow and a mitigation of lethal TBI. Starting infusion of aPC as late as 24 h after exposure to radiation was sufficient to mitigate radiation-induced mortality in these mice. These findings suggest that pharmacologic augmentation of the activity of the Thbd-aPC pathway by recombinant Thbd or aPC might offer a rational approach to the mitigation of tissue injury and lethality caused by ionizing radiation.


Cancer Research | 2008

Reciprocal relationship between O6-methylguanine-DNA methyltransferase P140K expression level and chemoprotection of hematopoietic stem cells.

Michael D. Milsom; Moran Jerabek-Willemsen; Chad E. Harris; Axel Schambach; Emily Broun; Jeff Bailey; Michael Jansen; David Schleimer; Kalpana Nattamai; Jamie Wilhelm; Amanda J. Watson; Hartmut Geiger; Geoffrey P. Margison; Thomas Moritz; Christopher Baum; Jürgen Thomale; David A. Williams

Retroviral-mediated delivery of the P140K mutant O(6)-methylguanine-DNA methyltransferase (MGMT(P140K)) into hematopoietic stem cells (HSC) has been proposed as a means to protect against dose-limiting myelosuppressive toxicity ensuing from chemotherapy combining O(6)-alkylating agents (e.g., temozolomide) with pseudosubstrate inhibitors (such as O(6)-benzylguanine) of endogenous MGMT. Because detoxification of O(6)-alkylguanine adducts by MGMT is stoichiometric, it has been suggested that higher levels of MGMT will afford better protection to gene-modified HSC. However, accomplishing this goal would potentially be in conflict with current efforts in the gene therapy field, which aim to incorporate weaker enhancer elements to avoid insertional mutagenesis. Using a panel of self-inactivating gamma-retroviral vectors that express a range of MGMT(P140K) activity, we show that MGMT(P140K) expression by weaker cellular promoter/enhancers is sufficient for in vivo protection/selection following treatment with O(6)-benzylguanine/temozolomide. Conversely, the highest level of MGMT(P140K) activity did not promote efficient in vivo protection despite mediating detoxification of O(6)-alkylguanine adducts. Moreover, very high expression of MGMT(P140K) was associated with a competitive repopulation defect in HSC. Mechanistically, we show a defect in cellular proliferation associated with elevated expression of MGMT(P140K), but not wild-type MGMT. This proliferation defect correlated with increased localization of MGMT(P140K) to the nucleus/chromatin. These data show that very high expression of MGMT(P140K) has a deleterious effect on cellular proliferation, engraftment, and chemoprotection. These studies have direct translational relevance to ongoing clinical gene therapy studies using MGMT(P140K), whereas the novel mechanistic findings are relevant to the basic understanding of DNA repair by MGMT.


Cell Reports | 2015

Stem cell specific mechanisms ensure genomic fidelity within HSCs and upon aging of HSCs

Bettina Moehrle; Kalpana Nattamai; Andreas Brown; Maria Carolina Florian; Marnie A. Ryan; Mona Vogel; Corinna Bliederhaeuser; Karin Soller; Daniel R. Prows; Amir Abdollahi; David Schleimer; Dagmar Walter; Michael D. Milsom; Peter J. Stambrook; Matthew H. Porteus; Hartmut Geiger

Whether aged hematopoietic stem and progenitor cells (HSPCs) have impaired DNA damage repair is controversial. Using a combination of DNA mutation indicator assays, we observe a 2- to 3-fold increase in the number of DNA mutations in the hematopoietic system upon aging. Young and aged hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) do not show an increase in mutation upon irradiation-induced DNA damage repair, and young and aged HSPCs respond very similarly to DNA damage with respect to cell-cycle checkpoint activation and apoptosis. Both young and aged HSPCs show impaired activation of the DNA-damage-induced G1-S checkpoint. Induction of chronic DNA double-strand breaks by zinc-finger nucleases suggests that HSPCs undergo apoptosis rather than faulty repair. These data reveal a protective mechanism in both the young and aged hematopoietic system against accumulation of mutations in response to DNA damage.


Cell Reports | 2017

Canonical Wnt Signaling Ameliorates Aging of Intestinal Stem Cells

Kodandaramireddy Nalapareddy; Kalpana Nattamai; R. Kumar; Rebekah Karns; Kathryn A. Wikenheiser-Brokamp; Leesa Sampson; Maxime M. Mahe; Nambirajan Sundaram; Mary-Beth Yacyshyn; Bruce R. Yacyshyn; Michael A. Helmrath; Yi Zheng; Hartmut Geiger

SUMMARY Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.


Methods of Molecular Biology | 2009

Quantification of genomic mutations in murine hematopoietic cells.

Hartmut Geiger; Schleimer David; Kalpana Nattamai; Vijg Jan

Maintaining the stability of the genome is critical to cell survival and normal cell growth. Genetic modification of hematopoietic cells might bear an inherent increased risk for the accumulation of DNA mutations. It frequently requires cultivation of the cells under super-physiological oxygen levels, which can result in increased oxidative damage, as well as under super-physiological concentrations of cytokines, which might interfere with DNA-damage checkpoint activation and by this means might result in an increased mutational load. We describe here a protocol for monitoring the frequency of DNA mutations in bone marrow cells post transduction or upon selection either in vitro or in vivo based on the lacZ-plasmid (pUR288) transgenic mouse (small blue mouse) mutation indicator strain.


Stem cell reports | 2017

The Spindle Assembly Checkpoint Is Required for Hematopoietic Progenitor Cell Engraftment

Andreas Brown; Johannes Pospiech; Karina Eiwen; Darren J. Baker; Bettina Moehrle; Vadim Sakk; Kalpana Nattamai; Mona Vogel; Ani Grigoryan; Hartmut Geiger

Summary The spindle assembly checkpoint plays a pivotal role in preventing aneuploidy and transformation. Many studies demonstrate impairment of this checkpoint in cancer cells. While leukemia is frequently driven by transformed hematopoietic stem and progenitor cells (HSPCs), the biology of the spindle assembly checkpoint in such primary cells is not very well understood. Here, we reveal that the checkpoint is fully functional in murine progenitor cells and, to a lesser extent, in hematopoietic stem cells. We show that HSPCs arrest at prometaphase and induce p53-dependent apoptosis upon prolonged treatment with anti-mitotic drugs. Moreover, the checkpoint can be chemically and genetically abrogated, leading to premature exit from mitosis, subsequent enforced G1 arrest, and enhanced levels of chromosomal damage. We finally demonstrate that, upon checkpoint abrogation in HSPCs, hematopoiesis is impaired, manifested by loss of differentiation potential and engraftment ability, indicating a critical role of this checkpoint in HSPCs and hematopoiesis.


PLOS Biology | 2018

Aging alters the epigenetic asymmetry of HSC division

M.Carolina Florian; Markus Klose; Mehmet Sacma; Jelena Jablanovic; Luke Knudson; Kalpana Nattamai; Gina Marka; Angelika Vollmer; Karin Soller; Vadim Sakk; Nina Cabezas-Wallscheid; Yi Zheng; Medhanie A. Mulaw; Ingmar Glauche; Hartmut Geiger

Hematopoietic stem cells (HSCs) balance self-renewal and differentiation to maintain homeostasis. With aging, the frequency of polar HSCs decreases. Cell polarity in HSCs is controlled by the activity of the small RhoGTPase cell division control protein 42 (Cdc42). Here we demonstrate—using a comprehensive set of paired daughter cell analyses that include single-cell 3D confocal imaging, single-cell transplants, single-cell RNA-seq, and single-cell transposase-accessible chromatin sequencing (ATAC-seq)—that the outcome of HSC divisions is strongly linked to the polarity status before mitosis, which is in turn determined by the level of the activity Cdc42 in stem cells. Aged apolar HSCs undergo preferentially self-renewing symmetric divisions, resulting in daughter stem cells with reduced regenerative capacity and lymphoid potential, while young polar HSCs undergo preferentially asymmetric divisions. Mathematical modeling in combination with experimental data implies a mechanistic role of the asymmetric sorting of Cdc42 in determining the potential of daughter cells via epigenetic mechanisms. Therefore, molecules that control HSC polarity might serve as modulators of the mode of stem cell division regulating the potential of daughter cells.


Experimental Hematology | 2017

Loss of DEK induces radioresistance of murine restricted hematopoietic progenitors

Juana Serrano-Lopez; Kalpana Nattamai; Nicholas A. Pease; Miranda S. Shephard; Ashley Wellendorf; Mathieu Sertorio; Eric A. Smith; Hartmut Geiger; Susanne I. Wells; Jose A. Cancelas; Lisa M. Privette Vinnedge

Self-renewing hematopoietic stem cells and multipotent progenitor cells are responsible for maintaining hematopoiesis throughout an individuals lifetime. For overall health and survival, it is critical that the genome stability of these cells is maintained and that the cell population is not exhausted. Previous reports have indicated that the DEK protein, a chromatin structural protein that functions in numerous nuclear processes, is required for DNA damage repair in vitro and long-term engraftment of hematopoietic stem cells in vivo. Therefore, we investigated the role of DEK in normal hematopoiesis and response to DNA damaging agents in vivo. Here, we report that hematopoiesis is largely unperturbed in DEK knockout mice compared with wild-type (WT) controls. However, DEK knockout mice have fewer radioprotective units, but increased capacity to survive repeated sublethal doses of radiation exposure compared with WT mice. Furthermore, this increased survival correlated with a sustained quiescent state in which DEK knockout restricted hematopoietic progenitor cells (HPC-1) were nearly three times more likely to be quiescent following irradiation compared with WT cells and were significantly more radioresistant during the early phases of myeloid reconstitution. Together, our studies indicate that DEK functions in the normal hematopoietic stress response to recurrent radiation exposure.

Collaboration


Dive into the Kalpana Nattamai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Zheng

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Schleimer

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marnie A. Ryan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Deidre Daria

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge