Network


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

Hotspot


Dive into the research topics where Lisa M. Curtis is active.

Publication


Featured researches published by Lisa M. Curtis.


American Journal of Physiology-renal Physiology | 2011

Paracrine effects of mesenchymal stem cells in cisplatin-induced renal injury require heme oxygenase-1.

Abolfazl Zarjou; Junghyun Kim; Amie Traylor; Paul W. Sanders; József Balla; Anupam Agarwal; Lisa M. Curtis

Multipotent mesenchymal stem cells (MSC) have become a popular and promising therapeutic approach in many clinical conditions. MSC are beneficial in animal models of acute kidney injury (AKI), by mediating differentiation-independent paracrine properties, and have prompted ongoing clinical trials to evaluate the safety and efficacy of MSC. Heme oxygenase-1 (HO-1) is induced in response to stress including AKI and has important anti-apoptotic, anti-inflammatory, and proangiogenic properties in these settings. We therefore examined whether HO-1 plays a role in the beneficial effects of MSC in AKI. We isolated MSC from bone marrow of age-matched HO-1+/+ and HO-1-/- mice. Our studies indicate that while differentiation of MSC into osteo- and adipocytic lineages did not differ between cells isolated from HO-1+/+ and HO-1-/- mice, MSC from HO-1-/- mice had significantly lower angiogenic potential. Moreover, HO-1-/- MSC demonstrated reduced expression and secretion of several important growth and proangiogenic factors (stromal cell-derived factor-1, vascular endothelial growth factor-A, and hepatocyte growth factor) compared with MSC derived from HO-1+/+ mice. In addition, conditioned medium of HO-1+/+ MSC rescued functional and morphological changes associated with cisplatin-induced AKI, while the HO-1-/--conditioned medium was ineffectual. Our studies indicate that HO-1 plays an important role in MSC-mediated protection. The results expand understanding of the renoprotective effects of MSC and may provide novel strategies to better utilize MSC in various disease models.


Journal of The American Society of Nephrology | 2015

Heme Oxygenase-1 Regulates Myeloid Cell Trafficking in AKI

Travis D. Hull; Ahmed I. Kamal; Ravindra Boddu; Subhashini Bolisetty; Lingling Guo; Cornelia C. Tisher; Sunil Rangarajan; Bo Chen; Lisa M. Curtis; James F. George; Anupam Agarwal

Renal ischemia-reperfusion injury is mediated by a complex cascade of events, including the immune response, that occur secondary to injury to renal epithelial cells. We tested the hypothesis that heme oxygenase-1 (HO-1) expression, which is protective in ischemia-reperfusion injury, regulates trafficking of myeloid-derived immune cells in the kidney. Age-matched male wild-type (HO-1(+/+)), HO-1-knockout (HO-1(-/-)), and humanized HO-1-overexpressing (HBAC) mice underwent bilateral renal ischemia for 10 minutes. Ischemia-reperfusion injury resulted in significantly worse renal structure and function and increased mortality in HO-1(-/-) mice. In addition, there were more macrophages (CD45(+) CD11b(hi)F4/80(lo)) and neutrophils (CD45(+) CD11b(hi) MHCII(-) Gr-1(hi)) in HO-1(-/-) kidneys than in sham and HO-1(+/+) control kidneys subjected to ischemia-reperfusion. However, ischemic injury resulted in a significant decrease in the intrarenal resident dendritic cell (DC; CD45(+)MHCII(+)CD11b(lo)F4/80(hi)) population in HO-1(-/-) kidneys compared with controls. Syngeneic transplant experiments utilizing green fluorescent protein-positive HO-1(+/+) or HO-1(-/-) donor kidneys and green fluorescent protein-negative HO-1(+/+) recipients confirmed increased migration of the resident DC population from HO-1(-/-) donor kidneys, compared to HO-1(+/+) donor kidneys, to the peripheral lymphoid organs. This effect on renal DC migration was corroborated in myeloid-specific HO-1(-/-) mice subjected to bilateral ischemia. These mice also displayed impaired renal recovery and increased fibrosis at day 7 after injury. These results highlight an important role for HO-1 in orchestrating the trafficking of myeloid cells in AKI, which may represent a key pathway for therapeutic intervention.


American Journal of Physiology-renal Physiology | 2008

Contribution of intrarenal cells to cellular repair after acute kidney injury: subcapsular implantation technique.

Lisa M. Curtis; Sifeng Chen; Bo Chen; Anupam Agarwal; Christopher A. Klug; Paul W. Sanders

The kidney is capable of regeneration following injury, particularly following acute insults. Although the mechanisms underlying cellular regeneration are incompletely understood, emerging evidence suggests a role for cells of renal origin in the repair and replacement of damaged renal tubule cells. The overall hypothesis of this study is that native kidney cells that reside in a niche in the kidney provide robust contribution to the repair of kidney tubules following injury. To test this hypothesis, we utilized a model of renal ischemia-reperfusion injury that results in extensive morphological changes, particularly in the outer medulla. Renal tissue obtained from mice constitutively expressing Escherichia coli beta-galactosidase (ROSA26) was dissected from the cortex, outer medulla, or papilla and implanted under the renal capsule of the injured mice. Mice were allowed to recover for 7 days. Sections through the injured kidney demonstrated the presence of implant-derived cells in renal tubules in the outer medulla. The implanted renal region that exhibited the most robust response was the papilla, whereas tissue pieces from the cortex and outer medulla showed less contribution to recipient renal tubules. These results provide proof-of-principle evidence that renal-derived reparative cells reside in all regions of the kidney, perhaps more predominantly in the renal papilla. A greater understanding of the cell biology of renal repair by native kidney cells will provide further insight into the design of novel therapies in acute kidney injury, and the subcapsular implant technique described in this study may offer unique advantages to evaluate renal repair mechanisms.


American Journal of Physiology-renal Physiology | 2017

Unique sex- and age-dependent effects in protective pathways in acute kidney injury

Ravindra Boddu; Chunlan Fan; Sunil Rangarajan; Bhuvana Sunil; Subhashini Bolisetty; Lisa M. Curtis

Sex and age influence susceptibility to acute kidney injury (AKI), with young females exhibiting lowest incidence. In these studies, we investigated mechanisms which may underlie the sex/age-based dissimilarities. Cisplatin (Cp)-induced AKI resulted in morphological evidence of injury in all groups. A minimal rise in plasma creatinine (PCr) was seen in Young Females, whereas in Aged Females, PCr rose precipitously. Relative to Young Males, Aged Males showed significantly, but temporally, comparably elevated PCr. Notably, Aged Females showed significantly greater mortality, whereas Young Females exhibited none. Tissue KIM-1 and plasma NGAL were significantly lower in Young Females than all others. IGFBP7 levels were modestly increased in both Young groups. IGFBP7 levels in Aged Females were significantly elevated at baseline relative to Aged Males, and increased linearly through day 3, when these levels were comparable in both Aged groups. Plasma cytokine levels similarly showed a pattern of protective effects preferentially in Young Females. Expression of the drug transporter MATE2 did not explain the sex/age distinctions. Heme oxygenase-1 (HO-1) levels (~28-kDa species) showed elevation at day 1 in all groups with highest levels seen in Young Males. Exclusively in Young Females, these levels returned to baseline on day 3, suggestive of a more efficient recovery. In aggregate, we demonstrate, for the first time, a distinctive pattern of response to AKI in Young Females relative to males which appears to be significantly altered in aging. These distinctions may offer novel targets to exploit therapeutically in both females and males in the treatment of AKI.


Journal of The American Society of Nephrology | 2016

Heme Oxygenase-1 Gene Polymorphisms—Toward Precision Medicine for AKI

Lisa M. Curtis; Anupam Agarwal

Heme oxygenase-1 (HO-1) is a 32-kD microsomal enzyme that serves as the rate-limiting step in the breakdown of heme, releasing iron, carbon monoxide, and biliverdin. Using pharmacologic and genetic approaches, the induction of HO-1 is a protective response in renal and nonrenal settings of tissue


Journal of Cancer Science & Therapy | 2014

Ascorbic Acid in Cancer: A Renewed Hope?

Sunil Rangarajan; Bhuvana Sunil; Lisa M. Curtis

Ascorbic acid (AA), long known to treat scurvy, has had debatable use as an anti-neoplastic drug in the past. However recent in vitro and in vivo studies have revealed previously unexplored mechanisms through which AA selectively damages cancer cells without causing damage to normal cells. In view of newly emerging evidence, many clinical trials have been designed to study these effects in patients with different types of cancers. Promising results from these initial trials are giving renewed hope to the use of AA as an adjuvant to the conventional chemotherapeutic drugs to treat cancer, to alleviate toxicity from the treatment and to reduce patient morbidity.


American Journal of Physiology-renal Physiology | 2000

Heme oxygenase-1 gene ablation or expression modulates cisplatin-induced renal tubular apoptosis

Fumie Shiraishi; Lisa M. Curtis; Leigh Truong; Kenneth D. Poss; Gary A. Visner; Kirsten Madsen; Harry S. Nick; Anupam Agarwal


Journal of Clinical Investigation | 2012

Mechanism and prevention of acute kidney injury from cast nephropathy in a rodent model

Wei-Zhong Ying; Christopher E. Allen; Lisa M. Curtis; Kristal J. Aaron; Paul W. Sanders


American Journal of Physiology-renal Physiology | 2007

Detection of early changes in renal function using 99mTc-MAG3 imaging in a murine model of ischemia-reperfusion injury

John Roberts; Bo Chen; Lisa M. Curtis; Anupam Agarwal; Paul W. Sanders; Kurt R. Zinn


Kidney International | 2007

HOpe for contrast-induced acute kidney injury

Lisa M. Curtis; Anupam Agarwal

Collaboration


Dive into the Lisa M. Curtis's collaboration.

Top Co-Authors

Avatar

Anupam Agarwal

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Paul W. Sanders

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Sunil Rangarajan

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Bhuvana Sunil

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Bo Chen

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Chunlan Fan

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ravindra Boddu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Subhashini Bolisetty

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Abolfazl Zarjou

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ahmed I. Kamal

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge